WO2009141827A2 - Conjugué d'un polymère, d'un agent anti-angiogenèse et d'une fraction de ciblage et utilisations de ce conjugué pour traiter des maladies angiogéniques osseuses - Google Patents

Conjugué d'un polymère, d'un agent anti-angiogenèse et d'une fraction de ciblage et utilisations de ce conjugué pour traiter des maladies angiogéniques osseuses Download PDF

Info

Publication number
WO2009141827A2
WO2009141827A2 PCT/IL2009/000511 IL2009000511W WO2009141827A2 WO 2009141827 A2 WO2009141827 A2 WO 2009141827A2 IL 2009000511 W IL2009000511 W IL 2009000511W WO 2009141827 A2 WO2009141827 A2 WO 2009141827A2
Authority
WO
WIPO (PCT)
Prior art keywords
conjugate
tnp
bone
alendronate
backbone
Prior art date
Application number
PCT/IL2009/000511
Other languages
English (en)
Other versions
WO2009141827A3 (fr
Inventor
Ronit Satchi-Fainaro
Ehud Segal
Jindrich Kopecek
Pavla Kopeckova
Huaizhong Pan
Original Assignee
Ramot At Tel Aviv University Ltd.
University Of Utah
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ramot At Tel Aviv University Ltd., University Of Utah filed Critical Ramot At Tel Aviv University Ltd.
Priority to US12/993,855 priority Critical patent/US8703114B2/en
Priority to CN200980128588.6A priority patent/CN102105157B/zh
Priority to EP09750279.3A priority patent/EP2300021A4/fr
Publication of WO2009141827A2 publication Critical patent/WO2009141827A2/fr
Publication of WO2009141827A3 publication Critical patent/WO2009141827A3/fr
Priority to US14/242,901 priority patent/US20140212357A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/58Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. poly[meth]acrylate, polyacrylamide, polystyrene, polyvinylpyrrolidone, polyvinylalcohol or polystyrene sulfonic acid resin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/74Synthetic polymeric materials
    • A61K31/785Polymers containing nitrogen
    • A61K31/787Polymers containing nitrogen containing heterocyclic rings having nitrogen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/0019Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules
    • A61K49/0021Fluorescence in vivo characterised by the fluorescent group, e.g. oligomeric, polymeric or dendritic molecules the fluorescent group being a small organic molecule
    • A61K49/0041Xanthene dyes, used in vivo, e.g. administered to a mice, e.g. rhodamines, rose Bengal
    • A61K49/0043Fluorescein, used in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0054Macromolecular compounds, i.e. oligomers, polymers, dendrimers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis

Definitions

  • the present invention in some embodiments thereof, relates to chemical conjugates and their use in therapy and diagnosis and, more particularly, but not exclusively, to chemical conjugates of a polymer, an anti-angiogenesis agent and a targeting moiety, which are useful, for example, in the treatment and monitoring of bone related diseases and disorders such as bone cancer and bone metastases.
  • Osteosarcoma is the most common type of primary bone cancer and classified as a malignant mesenchymal neoplasm in which the tumor directly produces defective osteoid (immature bone). It is a highly vascular and extremely destructive malignancy that most commonly arises in the metaphyseal ends of long bones.
  • Prostate cancer is the most common cancer of males in industrialized countries and the second leading cause of male cancer mortality. Mortality in these patients is not due to primary tumor growth, but rather due to complications caused by metastases to vital organs. Prostate cancer predominantly metastasizes to bone, but other organ sites are affected including the lung, liver, and adrenal gland. Breast cancer also often metastasizes to bones. Bone metastases incidence in patients with advanced metastatic disease is approximately 70 %. Bone metastases are associated with considerable skeletal morbidity, including severe bone pain, pathologic fracture, spinal cord or nerve root compressions, and hypercalcemia of malignancy. Chemotherapy agents, hormonal deprivation and bisphosphonates are the common treatments for advanced metastatic disease. However, with time, the disease progresses to a phase when the standard therapy fails to control the malignancy and furtherer progresses to a highly chemotherapy-resistant state.
  • Angiogenesis inhibitors such as TNP-470 [Folkman, J. Apmis 2004; 112: 496-507], its non-toxic N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer-conjugated form, caplostatin [Satchi-Fainaro et al. Cancer Cell 2005; 7: 251-261], and Avastin [Hurwitz et al. N Engl J Med 2004; 350: 2335-2342] are emerging as a new modality of anticancer drugs.
  • TKIs small molecule tyrosine kinase inhibitors
  • Inhibitors of mTOR represent a third, smaller category of antiangiogenic therapies with one currently approved agent (Torisel).
  • At least two other approved anti-angiogenic agents may indirectly inhibit angiogenesis through mechanisms that are not completely understood (Velcade, Celgene)
  • VEGF vascular endothelial growth factor
  • Novel targeted angiogenesis inhibitors for use with or without other antineoplastic agents have therefore been sought for.
  • a major impediment towards this effort has been the inability to determine therapeutic efficacy, the lack of reliable surrogate markers of tumor angiogenesis, and the complexity of interactions between multiple host cells and malignant cells involved in tumor angiogenesis, which may limit the use of a single anti-angiogenic agent.
  • Another significant obstacle is that the vast majority of clinically used anti-cancer and anti-angiogenic drugs are small molecules that exhibit a short half-life in the bloodstream and a high overall clearance rate. These low-molecular weight drugs diffuse rapidly into healthy tissues and are distributed evenly within the body. As a consequence, relatively small amounts of the drug reach the target site, and therapy is associated with low efficacy and severe side effects.
  • TNP-470 is a low molecular weight synthetic analogue of fumagillin, which is capable of selectively inhibiting endothelial growth //; vitro. In clinical trials, this drug was found to slow tumor growth in many patients with metastatic cancer and exhibited a promising efficacy when used in combination with conventional chemotherapy. However, at the doses required for tumor regression, many patients experienced neurotoxicity. Due to its dose-limiting neurotoxicity, no further clinical studies were conducted for using TNP-470 per se. It has been concluded that clinical uses of TNP- 470 should be performed with this agent being targeted to tumor tissue, in order to increase its site specificity and reduce side effects.
  • Water-soluble polymers such as N-(2-Hydroxypropyl) methacrylamide copolymers (HPMA) are biocompatible, non-immunogenic and non-toxic carriers that enable specific delivery into tumor tissue [Satchi-Fainaro et al. Nat Med 2004; 10: 255- 261]. These macromolecules do not diffuse through normal blood vessels but rather accumulate selectively in the tumor site because of the enhanced permeability and retention (EPR) effect. This phenomenon of passive diffusion through the hyperpermeable neovasculature and localization in the tumor interstitium is observed in many solid tumors for macromolecular agents and lipids.
  • EPR enhanced permeability and retention
  • Conjugation of anti-cancer drugs such as TNP-470 with copolymers, such as HPMA should enable selective targeting of these drugs to tumor tissue and thus reduce side effects. Furthermore, such copolymer-drug conjugates should restrict the passage through the blood brain barrier and would prolong the circulating half-life of the drugs, hence inhibiting the growth of tumor endothelial and epithelia cells by exposing the cells to the conjugated drugs in the circulation for a longer time compared to the free drugs.
  • An example of the favorable characteristics obtained by conjugation of an anti- angiogenesis agent such as TNP-470 to HPMA has been described by Satchi-Fainaro et al. in WO 03/086382.
  • BPs Bisphosphonates
  • alendronate are compounds with a chemical structure similar to that of inorganic pyrophosphate (PPi), an endogenous regulator of bone mineralization.
  • bisphosphonates are established as effective treatments in clinical disorders such as osteoporosis, Paget' s disease of bone, myeloma, and bone metastases.
  • Bisphosphonates such as zoledronic acid, have been shown to inhibit angiogenesis [Wood et al. J Pharmacol Exp Ther 2002; 302: 1055-1061].
  • the pharmacokinetic profile of bisphosphonates which exhibit a strong affinity to bone mineral under physiological conditions, their low toxicity and anti-angiogenic activity are advantageous for targeting to tumors confined to bony tissues.
  • Alendronate is considered potent for the treatment of bone related diseases and cancer-associated hypercalcemia.
  • bone targeting agents are oligopeptides of Aspartate.
  • Wang et al. describe fluorescein-labeled bone-targeted model conjugates for detection purposes bearing 1 % loading of D- Asps on HPMA copolymer [Wang et al. 2003 Bioconjug Chem 14:853- 859]. The bone-targeting potential of this conjugate was tested in vitro and in vivo and was found to selectively accumulate in bone tissue [Wang et al. 2006, MoI Pharm 3:717-725].
  • WO 2004/062588 teaches water soluble polymeric conjugates for bone targeted drug delivery with improved pharmacokinetics parameters and better water solubility of the loaded drugs.
  • the polymeric drug delivery systems taught by this application are based on hydroxypropyl methacrylamide (HPMA) conjugates of bone-targeting drugs such as alendronate and D-Asps together with a bone-related therapeutic agent.
  • HPMA hydroxypropyl methacrylamide
  • the loading of alendronate and D-Asps onto the HPMA copolymer was 0.494 mmol/gram and 0.762 mmol/gram respectively.
  • PK2 (FCE28069) is a HPMA copolymer-doxorubicin-galactosamine conjugate, which was designed as a treatment for hepatocellular carcinoma or secondary liver disease [Seymour et al. Journal of Clinical Oncology 2002; 20:1668-1676].
  • Doxorubicin is an anthracycline antibiotic with limited solubility in physiological fluids, and is a well established anti-neoplastic drug.
  • Galactosamine binds to the hepatic asialoglycoprotein receptor (ASGPR) thus serving as a specific hepatic targeting moiety.
  • ASGPR hepatic asialoglycoprotein receptor
  • the enzymatic degradable linker is a tetrapeptide spacer (Gly-Phe-Leu-Gly), designed for cleavage by lysosomal cathepsins.
  • Hruby et al. [Journal of Applied Polymer Science 2006; 101: 3192-3201] have prepared and synthesized novel polymeric drug-delivery systems designed for bone targeting of anti-neoplasties based on biocompatible HPMA copolymers containing hydroxybisphosphonate targeting moieties and the model drugs radiotherapeutics 125 I, imaging agent 111 In, or the anticancer drug Doxorubicin.
  • the percentage of hydroxybisphosphonate loaded onto the HPMA copolymers was in the range of 1.3-4 mol %.
  • the present inventors have further devised and successfully practiced a novel process for preparing the conjugates described herein, while obtaining a high load of alendronate in the polymer as well as a homogenous size distribution, i.e. low polydispersity, of the polymer.
  • This process can be beneficially performed in a controlled manner at 30 °C.
  • the present inventors have designed and successfully prepared and practiced a novel conjugate of a polymer (e.g., a N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer), an anti-angiogenesis agent (e.g., TNP-470) and a bone targeting agent being D-Asp8.
  • a polymer e.g., a N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer
  • HPMA N-(2-hydroxypropyl)methacrylamide
  • TNP-470 an anti-angiogenesis agent
  • a bone targeting agent being D-Asp8.
  • a conjugate comprising an N-(2-hydroxypropyl)methacrylamide)-derived polymeric backbone having attached thereto TNP-470 and alendronate, wherein a load of the alendronate in the polymer is greater than 3 mol %
  • the load of the alendronate in the polymer is greater than 5 mol %.
  • the load of the alendronate in the polymer is about 7 mol %.
  • at least one of the TNP-470 and the alendronate is attached to the polymer via a linker.
  • each of the TNP-470 and the alendronate is attached to the polymer via a linker.
  • the linker is a biodegradable linker.
  • the biodegradable linker is selected from the group consisting of a pH-sensitive linker and an enzymatically cleavable linker.
  • the biodegradable linker is an enzymatically cleavable linker.
  • the enzymatically cleavable linker is cleaved by an enzyme which is expressed in tumor tissues.
  • the enzymatically cleavable linker is cleaved by an enzyme which is overexpressed in tumor tissues.
  • the enzyme is selected from a group consisting of Cathepsin B, Cathepsin K, Cathepsin D, Cathepsin H, Cathepsin L, legumain, MMP-2 and MMP-9.
  • the enzyme is Cathepsin K.
  • the linker comprises an oligopeptide group containing from 2 to 10 amino acid residues.
  • the oligopeptide is -[Gly-Gly-Pro-Nle]-.
  • the TNP-470 is linked to the polymer or to the linker via a spacer.
  • the spacer has the formula G-
  • n is an integer from 1 to 4; and G and K are each independently selected from the group consisting of NH, O and S.
  • G and K are each NH and n is 2.
  • the alendronate is attached to the polymer via a linker that comprises -[Gly-Gly-Pro-Nle]-
  • the conjugate has the general formula II, as described herein.
  • the conjugate has the structure:
  • x is an integer that equals 70- 99.9 and y and w are each independently an integer that equals 0.01-15.
  • the conjugate further comprising a labeling agent.
  • the labeling agent is selected from the group consisting of a fluorescent agent, a radioactive agent, a magnetic agent, a chromophore, a bioluminescent agent, a chemiluminescent agent, a phosphorescent agent and a heavy metal cluster.
  • the labeling agent is Fluorescein isothiocyanate.
  • Such conjugate is having, for example, the following structure:
  • x is an integer that equals 70-99.9; and y, z and w are each independently an integer that equals 0.01-15.
  • a conjugate comprising a polymeric backbone having attached thereto an anti-angiogenesis agent and a bone targeting moiety, the bone targeting moiety being an oligopeptide of aspartic acid which comprises from 2 to 100 amino acid residues.
  • the oligopeptide comprises from 2 to 20 amino acid residues. According to some embodiments of the invention, the oligopeptide comprises 8 amino acid residues.
  • the aspartic acid is selected from the group consisting of D-aspartic acid and L-aspartic acid.
  • the aspartic acid is D-aspartic acid.
  • At least one of the anti- angiogenesis agent and the bone targeting moiety is attached to the polymeric backbone via a linker.
  • the linker is a biodegradable linker.
  • each of the anti-angiogenesis agent and the bone targeting moiety is attached to the polymeric backbone via a linker.
  • the polymeric backbone is derived from a polymer that has an average molecular weight that ranges from 100 Da to 800 kDa.
  • the polymeric backbone is derived from a polymer selected from the group consisting of dextran, a water soluble polyamino acid, a polyethylenglycol (PEG), a polyglutamic acid (PGA), a polylactic acid (PLA), a polylactic-co-glycolic acid (PLGA), a poly(D,L-lactide-c ⁇ -glycolide) (PLA/PLGA), a poly(hydroxyalkylmethacrylamide), a polyglycerol, a polyamidoamine (PAMAM), and a polyethylenimine (PEI).
  • the polymer is N-(2- hydroxypropyl)methacrylamide).
  • the anti-angiogenesis agent is TNP-470.
  • the biodegradable linker is selected from the group consisting of a pH-sensitive linker and an enzymatically- cleavable linker, as described herein.
  • a pharmaceutical composition comprising, as an active ingredient, any of the conjugates described herein and a pharmaceutically acceptable carrier.
  • the composition is packaged in a packaging material and identified in print, in or on the packaging material, for use in the treatment of a bone related disease or disorder.
  • the conjugate comprises a labeling agent, the composition being packaged in a packaging material and identified in print, in or on the packaging material, for use in monitoring a bone related disease or disorder.
  • the bone related disease or disorder is associated with angiogenesis.
  • a method of treating a bone related disease or disorder in a subject in need thereof comprising administering to the subject a therapeutically effective amount of any of the conjugates described herein.
  • a use of the conjugate as described hereinabove as a medicament is provided.
  • FIG. 1 presents a scheme illustrating the synthesis of an HPMA copolymer- ALN-TNP-470 conjugate according to some embodiments of the present invention.
  • FIGs. 2A-B present data showing the synergistic inhibitory effect of the combined treatment of alendronate and TNP-470 on proliferation of endothelial cells in vitro.
  • FIG. 2A presents comparative plots showing the effect of ALN (closed squares), TNP-470 (open squares), ALN+TNP-470 0.01 pM (closed triangles) and ALN+TNP- 470 10 ⁇ M (open triangles), on the proliferation of human umbilical vain endothelial cells (HUVEC), and demonstrating the synergistic effect of a combined alendronate and TNP-470 treatment.
  • FIG. 1 presents comparative plots showing the effect of ALN (closed squares), TNP-470 (open squares), ALN+TNP-470 0.01 pM (closed triangles) and ALN+TNP- 470 10 ⁇ M (open triangles), on the proliferation of human umbilical vain endothelial cells (HUVEC), and demonstrating the synergistic
  • 2B presents a classic isobologram of ALN-TNP-470 combination treatment at IC 70 (closed squares), IC 50 (closed triangles) and IC 3O (closed circles).
  • the dashed circles represent synergism areas of the combined ALN+TNP-470 treatment.
  • the tables represent the CI values of each IC of combination treatments I and II. Data represent mean ⁇ SD.
  • FIGs.3A-G present the 2-D chemical structure of fluorosceinated-HPMA copolymer-ALN-TNP-470 conjugates according to some embodiments of the present invention (FIG. 3A); a diagram showing FPLC detection of unbound HPMA-ALN- TNP470 conjugate in the samples, in the presence and absence of Hydroxyapitate, at selected time points (FIG. 3B); and plots showing the percentages of HPMA-ALN- TNP-470 conjugate bound to hydroxyapatite as a function of the elution time (FIG. 3C); size exclusion chromatography (SEC) profile of conjugate polymerized by the classical polymerization method (Polymerization I conjugate; FIG.
  • SEC size exclusion chromatography
  • FIG. 3D conjugate polymerized by RAFT polymerization
  • FIG. 3E conjugate polymerized by RAFT polymerization
  • FIG. 3F a graph showing the hydrodynamic diameter size distribution of the HPMA-ALN-TNP-470 polymerization I conjugate and polymerization II conjugate
  • FIG. 3G an image of the polymer I conjugate particles obtained
  • FIGs. 4A-S present confocal images showing the intracellular trafficking of FITC-labeled HPMA copolymer-ALN-TNP-470 conjugate, according to some embodiments of the present invention, in human umbilical vain endothelial cells (HUVEC) and Saos-2 human osteosarcoma cells.
  • Single XY plane imaging of the conjugate (green) with PI (red) nuclei staining showed cytoplasmatic accumulation of the conjugate.
  • Cellular uptake analysis of the conjugate by HUVEC cells FIG. 4A
  • 5.7 ⁇ m Z-stack of 28 slices FIG. 4B
  • XZ image slice show similar conjugate and nuclei focal plane localization (FIG.
  • FIG. 4C Multi-channel overlay of HUVEC (FIG. 4G) and Saos-2 cells (FIG. 4K) stained with phalloidin (red) for actin filaments (FIGs. 4D and 4H) and DAPI (blue) for nuclei (FIGs. 4F and 4J) 12 hours post incubation with the ALN-TNP-470 conjugate (green) (FIGs. 4E, 41), showing cellular localization of the conjugate mostly around the nuclei; the FITC-labeled conjugate (green) (FIGs. 4L and 4P) colocalized with clathrin-coated vesicles labeled with transferrin (red) (FIGs.
  • FIG. 5 present comparative plots demonstrating that ALN and TNP-470 retain their antiangiogenic effect when bound to the HPMA copolymer.
  • the percentage of average cell growth is similar in the presence of polymer-conjugated ALN and TNP- 470 in HUVEC cells (closed triangles), Saos-2 cells (closed squares) and MG-63-Ras (closed diamonds) compared with a combination of free ALN and free TNP-470 in HUVEC cells (open triangles), in Saos-2 cells (open squares) and MAG-63-Ras (open diamonds.
  • FIGs. 6A-C present the effect of HPMA copolymer-ALN-TNP-470 conjugate and free ALN + free TNP-470 on the ability of HUVEC to migrate towards vascular endothelial growth factor (VEGF) chemoattractant and the ability to form capillary-like tube structures.
  • FIG. 6A presents a bar graph showing that free (dotted bars) and polymer-conjugated (gray bars) ALN and TNP-470 inhibited VEGF induced HUVEC migration
  • FIG. 6B presents images of HUVEC exposed to the combination of free ALN + free TNP-470, and with HPMA copolymer ALN-TNP470 after 8 hours of incubation; and FIG.
  • FIG. 6C presents a bar graph showing the percentages of inhibition of HUVEC capillary-like tube structures by different concentrations of free (dotted bars) and polymer-conjugated (gray bars) ALN and TNP-470, compared with non-treated cells (black bar). Data represent mean ⁇ SD.
  • FIGs. 7A-G present data showing that HPMA copolymer-ALN-TNP-470 conjugate, according to some embodiments of the present invention, reduces vascular hyperpermeability in mouse skin capillaries and inhibits MG-63-Ras human osteosarcoma growth.
  • FIG. 7A presents representative images showing diminished Evans Blue dye uptake in skin patches of mice treated with free ALN+TNP-470 or HPMA copolymer-ALN-TNP-470 conjugate as compared to untreated control group. Scale bar represents 10 mm.
  • FIG. 7C presents representative intravital non-invasive fluorescence images of mCherry-labeled MG-63-Ras tumor-bearing nontreated mice (control) or treated with free ALN+TNP-470 or HPMA copolymer-ALN-TNP-470 conjugate.
  • FIG. 7D presents comparative plots showing antitumor effect of free ALN + TNP-470 (open triangles) or HPMA copolymer-ALN-TNP-470 conjugate (closed triangles) on MG-63-Ras human osteosarcoma tumor size compared to vehicle-treated group (closed squares) and dissected tumors images.
  • Scale bar represents 10 mm.
  • Data represent mean ⁇ s.d.
  • FIG. 7E presents representative images from whole-mount confocal microscopy of mCherry-labeled MG-63-Ras human osteosarcoma tumors dissected from mice treated with FITC-labeled HPMA copolymer-ALN-TNP-470 conjugate. Scale bar represents 25 ⁇ m.
  • FIG. 7F presents representative images of H & E and CD34 immunostaining of control, free ALN + TNP-470 combination, and HPMA copolymer-ALN-TNP-470 conjugate treated MG-63-Ras osteosarcomas inoculated s.c. in mice.
  • 7G presents representative images of dissected organs of mice treated with FITC-labeled HPMA copolymer-ALN-TNP-470 conjugate showing greater intensity of FITC-fluorescence spectrum (green; composed images of unmixed multispectral cubes) in bone tissues then in the spleen, heart, lungs, kidneys and liver. Images were taken using the CRJ MaestroTM intravital non-invasive fluorescence imaging system.
  • FIG. 8 presents a scheme illustrating the synthesis of two HPMA copolymer-D- Asps-TNP-470 conjugates (HPl and HP2) according to some embodiments of the present invention.
  • FIG. 9 presents a size exclusion chromatography (SEC) profile of two HPMA copolymer-D-Asps-TNP-470 conjugates (HPl and HP2).
  • FIG. 10 presents comparative plots showing effect of free TNP-470 (orange squares), compared with HPMA-D-Asps-TNP-470 conjugates HPl (black squares) and HP2 (blue squares) on the growth of HUVEC, demonstrating that TNP-470 retains its anti-angiogenic effect when bound to the HPMA copolymer.
  • FIGs. 11A-B present bar graphs showing the effect of the HPMA-D- Asp 8 -TNP- 470 conjugates, HPl (FIG. HA) and HP2 (FIG. HB), compared with free TNP-470, on the ability of HUVEC to migrate towards vascular endothelial growth factor (VEGF) chemoattractant, and demonstrating that free and polymer-conjugated TNP-470 inhibited VEGF-induced HUVEC migration to a similar extent.
  • VEGF vascular endothelial growth factor
  • FIGs. 12A-B present comparative plots demonstrating that ALN exhibits an anti-angiogenic effect in a dose dependent manner. Shown in FIG. 12A are plots of the percentage of average HUVEC cell growth as a function of free ALN concentrations (red empty squares) and ALN conjugated to HPMA concentrations ( 1 mol % loading; filled blue squares). Shown in FIG. 12A
  • FIGS. 12A are plots of the percentage of average aggressive type (E) Saos-2 human osteosarcoma cell growth as a function of free ALN concentrations (red filled circles); ALN conjugated to HPMA concentrations ( 1 mol % loading; filled blue triangles) as well as plots of the percentage of average dormant (D) type Saos-2 human osteosarcoma cell growth as a function of free ALN concentrations (empty filled circles) and ALN conjugated to HPMA ( 1 mol % loading; empty blue triangles) concentrations.
  • Solid and dashed lines represent the proliferation of cells in the presence (solid line) or absence (dashed line) of growth factors. Data represent mean ⁇ SD.
  • the present invention in some embodiments thereof, relates to chemical conjugates and their use in therapy and diagnosis and, more particularly, but not exclusively, to chemical conjugates of a polymer, an anti-angiogenesis agent and a targeting moiety, which are useful, for example, in the treatment and monitoring of bone related diseases and disorders such as bone cancer and bone metastases.
  • the principles and operation of the conjugates, compositions, use, methods and processes according to the invention may be better understood with reference to the drawings and accompanying descriptions.
  • the present inventors have now devised and successfully prepared and practiced novel conjugates of a copolymer having attached thereto an anti-angiogenesis agent and a bone targeting moiety. More specifically, but not exclusively, the present inventors have devised and successfully practiced novel processes of preparing such conjugates, in which the bone targeting moiety is alendronate or an oligoaspartate.
  • the present inventors have devised and successfully practiced novel processes of preparing such conjugates in which alendronate is present in a relatively high load within the copolymer.
  • alendronate and TNP- 470 can act in synergy in inhibition of angiogenesis.
  • the alendronate high-loaded conjugates described herein are therefore characterized as highly potent agents for treating bone-related diseases and disorders.
  • the present inventors have successfully prepared and practiced a novel polymeric conjugate of a N-(2- hydroxypropyl)methacrylamide (HPMA) copolymer, having attached thereto TNP-470 and the bone targeting agent, alendronate (ALN), wherein the TNP-470 and alendronate are conjugated to backbone units of the HPMA-derived polymeric backbone via biodegradable linkers and the mol percent of alendronate loaded onto the conjugate is higher than in currently known alendronate-polymer conjugates (e.g., is greater than 3 mol % of the polymeric conjugate).
  • HPMA N-(2- hydroxypropyl)methacrylamide
  • This polymeric conjugate exhibited an enhanced inhibition of angiogenesis and bone cancer as compared with un-conjugated, i.e., free, TNP-470 and ALN, administered at equivalent doses.
  • this HPMA copolymer-ALN ⁇ TNP470 conjugate was capable of binding to bone mineral (see, Figures 3B and 3C), and exhibited a superior activity, as compared to non-conjugated ALN and TNP-470, with regard to inhibition of osteosarcoma cell proliferation (see, Figure 5), inhibition of endothelial cell proliferation and migration (see, Figures 5 and 6A respectively), inhibition of endothelial cell ability to form capillary tube structures (see, Figure 6B-C), and in reducing vascular hypermeability and osteosarcoma tumor growth in-vivo (see, Figure 7).
  • the present inventors have further designed and successfully prepared and practiced a novel conjugate of a polymer (e.g., a N-(2-hydroxypropyl) methacrylamide (HPMA)-derived co-polymer) having attached thereto an anti-angiogenesis agent (e.g., TNP-470) and a bone targeting agent being D-Asp8.
  • a polymer e.g., a N-(2-hydroxypropyl) methacrylamide (HPMA)-derived co-polymer
  • HPMA N-(2-hydroxypropyl) methacrylamide
  • a polymeric conjugate comprising an N-(2-hydroxypropyl)methacrylamide)- derived polymeric backbone having attached thereto TNP-470 and alendronate, wherein a load of the alendronate in the polymeric conjugate is greater than 3 mol %.
  • N-(2-hydroxypropyl)methacrylamide (HPMA) polymers are a class of water- soluble synthetic polymeric carriers that have been extensively characterized as biocompatible, non-immunogenic and non-toxic. HPMA polymers can be tailored through relatively simple chemical modifications, in order to regulate their respective drug and targeting moiety content.
  • HPMA conjugates are characterized by a limited diffusion and/or extravasation through normal blood vessels, due to the high molecular weight and hydrodynamic diameter thereof, and therefore accumulate selectively at the tumor site, which is characterized by leaky blood vessels having abnormal form and architecture and wide fenestrations, pores and vesicular vacuolar organelles (VVO).
  • VVO vesicular vacuolar organelles
  • Conjugating drugs to polymers such as HPMA is also expected to restrict the passage of the conjugate through the blood brain barrier, thus prolonging the circulating half-life of the drugs and abrogating neurotoxicity associated with many chemotherapeutic and anti- angiogenic drugs.
  • a polymeric conjugate comprising an N-(2-hydroxypropyl)methacrylamide)- derived polymeric backbone having attached thereto TNP-470 and alendronate, that has a molecular weight higher than 10 kDa typically exhibit an EPR effect, as described herein, while polymeric substances that have a molecular weight of 100 kDa and higher have relatively long half-lives in plasma and an inefficient renal clearance. Accordingly, a molecular weight of the polymeric conjugate can be determined while considering the half-life in plasma, the renal clearance, and the accumulation in the tumor of the conjugate. The molecular weight of the polymeric conjugate can be controlled, at least to some extent, by the degree of polymerization (or co- polymerization).
  • the conjugate described herein has a MW that ranges from 100 Daltons to SOO kDa. In some embodiments, the conjugate described herein has a MW that ranges from 10 kDa to 800 kDa. In some embodiments, the conjugate has a MW that ranges from 10 kDa to 60 kDa.
  • the conjugates described herein comprise an HPMA polymeric backbone comprised of N-(2- hydroxypropyl)methacrylamide-derived backbone units (a polymeric backbone formed by polymerizing N-(2-hydroxypropyl)methacrylamide monomers), whereby TNP-470 molecules are attached to a portion of these backbone units and alendronate molecules are attached to another portion of these backbone units, as described herein.
  • Those backbone units within the polymeric backbone that are not linked to another moiety e.g., TNP-470, alendronate, or any of the other moieties described herein
  • free or non-functionalized backbone units are referred to herein as "free" or "non-functionalized” backbone units.
  • polymeric backbone in the conjugates described herein is composed of some backbone units that have alendronate attached thereto, some backbone units that have TNP-470 attached thereto, and optionally some free backbone units, these conjugates represent HPMA-derived co-polymers.
  • TNP-470 is a potent anti-angiogenesis agent. Its use as a free drug has been limited by its low solubility and dose-dependent neurotoxicity.
  • anti-angiogenesis agent which is also referred to herein interchangeably as “anti-angiogenic agent” or “angiogenesis inhibitor”, describes an agent having the ability to (a) inhibit endothelial cell proliferation or migration; (b) kill proliferating endothelial cells; and/or (c) inhibit the formation of new blood vessels in a tissue.
  • alendronate (4-amino-l-hydroxybutylidene) bisphosphonic acid) is a bisphosphonate which exhibits a strong affinity to bone minerals under physiological conditions.
  • alendronate also encompasses any pharmaceutically acceptable salts, solvates and/or hydrates thereof, as defined hereinafter.
  • alendronate exhibits anti-angiogenesis activity in a dose dependent manner.
  • Cheng et al. have shown that alendronate reduces the mRNA level and cellular level of Matrix metalloproteinase-2 (MMP-2) enzyme in osteosarcoma cell lines in a time and dose-dependent manner [Cheng et al.
  • MMP-2 Matrix metalloproteinase-2
  • the anti-angiogenesis activity of alendronate is dose dependent, as assessed by the extent of HUVEC and
  • Saos-2 human osteosarcoma cell line proliferation inhibition whereby the extent of inhibition is proportional to the alendronate concentrations, i.e., at higher alendronate concentration a stronger anti-angiogenesis activity could be observed.
  • Conjugating alendronate and an anti-angiogenesis agent to polymers is highly beneficial for producing an agent that is characterized by both selectivity, due to the EPR effect attributed to the polymer, and the bone-targeting effect attributed to the alendronate, and a potent therapeutic activity, due to the presence of a potent anti- angiogenesis agent.
  • conjugates which are further characterized by a high load of alendronate, as described herein, are even more potent, due to the dual targeting and anti-angiogenesis effect that can be potentially exhibited by the alendronate.
  • the conjugates described herein are characterized by an alendronate loading which is higher than 3 mol %.
  • loading or simply “load”, and any grammatical diversion thereof, is used to describe the amount of an agent that is attached to the polymeric backbone of the conjugates described herein, and is represented herein by the mol % of this agent in the conjugate, as defined hereinafter.
  • mol % describes the number of moles of an attached moiety per 1 mol of the polymeric conjugate, multiplied by 100.
  • a 3 mol % load of alendronate describes a polymeric conjugate composed of 100 backbone HPMA units, whereby 3 HPMA backbone units have alendronate-containing monomer units attached thereto, and the other 97 HPMA backbone units are either free or have other agents attached thereto.
  • the load of alendronate can be, for example, 3, 3.1, 3.2, 3.3, 3.4, 3.5, 4, 4.5, 5, 5.5, 6, 6.5, 7, 7.5, 8, 8.5, 9, 9.5 and even as high as 10 mol %.
  • the load of the alendronate in the polymer is greater than 5 mol %.
  • the load of the alendronate in the polymer is about 7 mol %.
  • the phrase "about” describes ⁇ 1%.
  • the phrase “about 7 mol %” describes a mol percentage ranging between 6 mol % and 8 mol %, or between 6.5 mol % and 7.5 mol %.
  • this phrase encompasses 6.5 mol %, 6.6 mol %, 6.7 mol %, 6.8 mol %, 6.9 mol %, 7 mol %, 7.1 mol %, 7.2 mol %, 7.3 mol %, 7.4 mol % and 7.5 mol %.
  • alendronate can exhibit both a bone targeting effect and an anti-angiogenesis effect.
  • a combined therapy of TNP-470 and alendronate results in a synergistic anti-angiogenic activity, when alendronate is utilized at a high concentration.
  • the anti-angiogenesis activity of alendronate and TNP-470 when administered together, as demonstrated by their inhibitory effect on the proliferation of endothelial cells, was superior to the cumulative anti-angiogenesis activity of each agent when administered alone (see, Figure 2).
  • This synergistic activity could be observed only at high alendronate concentration whereas at low alendronate concentration (lower than 100 nM), no synergistic activity could be detected.
  • alendronate exhibits a dose-dependent anti-angiogenesis activity, and further since it is shown herein that alendronate and TNP-470 can act in synergy in inhibition of angiogenesis, such high-loaded conjugates can be beneficially utilized in the treatment of bone-related diseases and disorders such as those conditions that are associated with angiogenesis.
  • the TNP-470 and alendronate that are attached to the polymer in the conjugates described herein act in synergy.
  • “Synergy” or “synergistic activity” is therefore often determined when a value representing an effect of a combination of two active agents is greater than the sum of the same values obtained for each of these agents when acting alone.
  • a synergy between two anti-angiogenesis agents may be determined by methods well known in the art.
  • the alendronate and the TNP -470 can each be linked to the polymeric backbone directly, or indirectly, through a linker moiety (also referred to herein as a linker, a linker group or a linking group), whereby, in some embodiments, the direct/indirect linkage is designed as being cleavable at conditions characterizing the environment of a desired bodily site (e.g., by certain enzymes or pH), as detailed hereinbelow.
  • a linker moiety also referred to herein as a linker, a linker group or a linking group
  • At least one of the TNP-470 and the alendronate is attached to the polymeric backbone via a linker.
  • each of the TNP-470 and the alendronate is attached to the polymeric backbone via a linker.
  • the linker linking the TNP-470 to the polymer and the linker linking the alendronate to the polymeric backbone may be the same or different.
  • the linker described herein refers to a chemical moiety that serves to couple the
  • TNP-470 and/or the alendronate to the polymeric backbone while not adversely affecting either the targeting function of the alendronate or the therapeutic effect of the alendronate and/or the TNP-470.
  • the linker is a biodegradable linker.
  • biodegradable linker describes a linker that is capable of being degraded, or cleaved, when exposed to physiological conditions.
  • physiological conditions can be, for example, pH, a certain enzyme, and the like.
  • the linker is capable of being cleaved by pre-selected cellular enzymes, for instance, those found in osteoblasts, osteoclasts, lysosomes of cancerous cells or proliferating endothelial cells.
  • an acid hydrolysable linker could comprise an ester or amide linkage and be for instance, a cis-aconityl linkage.
  • Such linkers further enhance the therapeutic activity and reduced toxicity of the conjugates described herein, by allowing the release of the anti-angiogenesis drug and/or the alendronate only at the desired bodily site.
  • the biodegradable linker is a pH- sensitive linker or an enzymatically-cleavable linker.
  • a pH-sensitive linker comprises a chemical moiety that is cleaved or degraded only when subjected to a certain pH condition, such as acidic pH (e.g., lower than 7), neutral pH (6.5-7) or basic pH (higher than 7).
  • Such a linker may, for example, be designed to undergo hydrolysis under acidic or basic conditions, and thus, the conjugate remains intact and does not release the agents attached to the polymer in the body, until its reaches a physiological environment where a pH is either acidic or basic, respectively.
  • Exemplary pH-sensitive linkers include, but are not limited to hydrazone bond, ester (including orthoesther) bonds, amide bonds of a cis-aconytil residue, a trityl group, acetals, ketals, Alanine ester, Gly-ester and a -[Gly-Phe-Gly]- moiety.
  • the biodegradable linker is an enzymatically-cleavable linker.
  • Such a linker is typically designed so as to include a chemical moiety, typically, but not exclusively, an amino acid sequence, that is recognized by a pre-selected enzyme. Such an amino acid sequence is often referred to in the art as a "recognition motif".
  • a conjugate comprising such a linker typically remains substantially intact in the absence of the pre-selected enzyme, and hence does not cleave or degrade so as to the release the agent attached thereto until it reaches an environment where this enzyme is present at a substantial concentration.
  • the enzymatically cleavable linker is cleaved by an enzyme which is expressed in tumor tissues.
  • a conjugate comprising such a linker ensures, for example, that a substantial amount of the conjugated TNP-470 is released from the conjugate only at the tumor tissue, thus reducing the side effects associated with the non-selective administration of the drug.
  • the enzymatically cleavable linker is cleaved by an enzyme which is overexpressed in tumor tissues.
  • exemplary enzymes which are suitable for use in the context of these embodiments include, but are not limited to, Cathepsin B, Cathepsin K, Cathepsin D,
  • Suitable linkers include, but are not limited to, alkyl chains; alkyl chains optionally substituted with one or more substituents and in which one or more carbon atoms are optionally interrupted by a nitrogen, oxygen and/or sulfur heteroatom.
  • linkers include amino acids and/or oligopeptides.
  • alkyl chains and/or oligopeptides can optionally be functionalized so as allow their covalent binding to the moieties linked thereby (e.g., the polymeric backbone and the alendronate, the polymeric backbone and the TNP-470).
  • a functionalization may include incorporation or generation of reactive groups that participate in such covalent bindings.
  • the linker is a biodegradable oligopeptide which contains, for example, from 2 to 10 amino acid residues.
  • the linker is a Cathepsin K-cleavable linker.
  • Cathepsin K is a lysosomal cysteine protease involved in bone remodeling and resorption and is predominantly expressed in osteoclasts. Its expression is stimulated by inflammatory cytokines that are released after tissue injury and in bone neoplasms [Pan et al. 2006, J Drug Target 14:425-435; Husmann et al. 2008, MoI Carcinog 47: 66-73].
  • An exemplary linker having Cathepsin K cleavable sites is -[Gly-Gly-Pro- NIe]-.
  • the linker comprises the amino acid sequence -[GIy-GIy- Pro-Nle]-.
  • the linker consists of the amino acid sequence -[GIy-GIy- Pro-Nle]-.
  • Cathepsin K cleavable sites are those that include a -[Gly-Pro-Nle]- moiety.
  • Another example include [-Gly-Gly-NH-C ⁇ -Gly-Pro-Nle]-.
  • a Cathepsin K cleavable linker being -[Gly-Gly-Pro-Nle]- was used, linking both alendronate and TNP-470 to the HPMA polymeric backbone (see, Figure 1).
  • a HPMA copolymer-ALN-TNP-470 conjugate comprising such linker moieties successfully inhibited proliferation of endothelial cells as well as Saos-2 and MG-63-Ras human osteosarcoma cells.
  • the involvement of Cathepsin K in the release of the TNP-470 and alendronate from the polymer could be deduced from the reduced activity of the conjugate when incubated together with a cathepsin K inhibitor whereby the conjugate inhibited the proliferation of HUVEC at a 4-logs higher concentration in the presence of cathepsin K inhibitor III than in its absence (see, Figure 5).
  • An oligopeptide linker which contains the pre-selected amino acid sequence can also be constructed such that the recognition motif is repeated several times within the linker, to thereby enhance the selective release of the attached agent.
  • recognition motif of the same or different enzymes can also be incorporated within the linker.
  • the linker may comprise multiple pH sensitive bonds or moieties. Linkers comprising such multiple cleavable sites can enhance the selective release of the anti-angiogenesis agent at the desired bodily site, thereby reducing adverse side effects, and further enhance the relative concentration of the released drug at the bodily site when it exhibits its activity.
  • the bond linking these moieties can also be biodegradable, for example, an enzymatically-cleavable bond or a pH-sensitive bond (e.g., an acid- hydrolyzable bond).
  • a bond can be formed upon functionalizing backbone units of the polymeric backbone, the alendronate and/or the TNP-470, so as to include compatible reactive groups for forming the desired bond.
  • the TNP-470 is linked to the polymer or to the linker via a spacer.
  • the alendronate is linked to the polymer or to the linker via a spacer.
  • the spacers can be the same or different.
  • spacer as used herein describes a chemical moiety that is covalently attached to, and interposed between, the polymeric backbone and the linker, the TNP- 470 and/or the alendronate, thereby forming a bridge-like structure between the polymeric backbone and the linker, the TNP-470 and/or the alendronate.
  • the spacer does not actively participate in a biological process, and is present in the conjugate for the purpose of facilitating its synthesis and/or improving its performance in terms of, for example, steric considerations, as is detailed hereinbelow.
  • Suitable spacers include, but are not limited to, alkylene chains, optionally substituted by one or more substituents and which are optionally interrupted by one or more nitrogen, oxygen and/or sulfur heteroatom.
  • Other suitable spacers include amino acids and amino acid sequences, optionally functionalized with one or more reactive groups for being coupled to the polymeric backbone, TNP-470 and/or alendronate.
  • the spacer has the formula G-(CH 2 )n-K, wherein n is an integer from 1 to 10; and G and K are each a reactive group, as defined herein, such as, for example, NH, O or S. In some embodiments, G and K are each NH and n is 2.
  • the spacer is an amino acid sequence, optionally an inert amino acid sequence (namely, does not affect the affinity or selectivity of the conjugate).
  • a spacer can be utilized for elongating or functionalizing the linker.
  • a spacer is utilized for enabling a more efficient and simpler attachment of the alendronate and/or the TNP-470 to the polymeric backbone or linker, in terms of steric considerations (renders the site of the polymeric backbone to which coupling is effected less hindered) or chemical reactivity considerations (adds a compatible reactive group to the site of the polymeric backbone to which coupling is effected).
  • the spacer may contribute to the performance of the resulting conjugate.
  • the spacer may render an enzymatically cleavable linker less sterically hindered and hence more susceptible to enzymatic interactions.
  • the spacer may also be used in order to attach other agents (e.g., a labeling agent, as described hereinbelow) to the conjugate.
  • the spacer may be varied in length and in composition, depending on steric and chemical considerations, and may be used to space the TNP-470 and alendronate form the polymeric backbone and/or the linker.
  • the present inventors have successfully synthesized a conjugate wherein the TNP-470 is linked to the HPMA polymeric backbone via a cathepsin K cleavable linker being-[Gly-Gly-Pro-Nle]- and a spacer being -NH-(CH 2 ) 2 -NH- (see, Figure 1).
  • the degree of loading of the TNP-470 and alendronate may be expressed as mole %, as defined herein.
  • the optimal degree of loading of TNP-470 is determined empirically based on the desired properties of the conjugate (e.g., water solubility, therapeutic efficacy, pharmacokinetic properties, toxicity and dosage requirements), and synthetic considerations (e.g., the amount of the drug that can be attached to the backbone units in a certain synthetic pathway).
  • desired properties of the conjugate e.g., water solubility, therapeutic efficacy, pharmacokinetic properties, toxicity and dosage requirements
  • synthetic considerations e.g., the amount of the drug that can be attached to the backbone units in a certain synthetic pathway.
  • the loading of TNP-470 in the polymer is greater than 1 mol %. In some embodiments, the loading of the TNP-470 in the conjugate ranges from
  • 1 mol % to 90 mol % from 1 mol % to 50 mol %, from 1 mol % to 20 mol %, from 1 mol % to 10 mol %, or from 1 mol % to 5 mol %.
  • the number of backbone units in the polymeric backbone having TNP-470 attached thereto is defined herein as "y"
  • the number of backbone units in the polymeric backbone having alendronate attached thereto is herein defined as "w”
  • the number of free backbone units in the polymeric backbone (which are not bound to an additional moiety) is herein defined as "x”.
  • the conjugate described herein can be represented by the general formula II:
  • x is an integer having a value such that x/(x+y+w) multiplied by 100 is in the range of from 0.01 to 99.9
  • y is an integer having a value such that y/(x+y+w) multiplied by 100 is in the range of from 0.01 to 99.9
  • w is an integer having a value such that w/(x+y+w) multiplied by 100 is in the range of from 3 to 99
  • B is TNP-470
  • D is alendronate
  • each of L 1 and L 2 is independently the linker, as described herein.
  • the conjugate has the following structure:
  • x/(x+y+w) multiplied by 100 may be 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or
  • y/(x+y+w) multiplied by 100 may be 0.01, 0.02, 0.03, 0.04, 0.05, 0.1, 0.2, 0.3,
  • 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 or 15; and w/(x+y+w) multiplied by 100 may be 5, 5.2, 5.4, 5.6, 5.8, 6, 6.2, 6.4, 6.6, 6.8, 7, 7.2,
  • w/(x+y+w) multiplied by 100 is 7.
  • the conjugate described herein further comprises a labeling agent attached thereto.
  • the labeling agent is attached to a portion of the backbone units that do not have the TNP-470 or the alendronate attached thereto.
  • the number of backbone units in the polymeric backbone having the labeling agent attached thereto is defined as V, as shown in the general Formula hereinbelow.
  • the attachment of a labeling agent to the conjugate enables utilizing these conjugates for monitoring bone related disease or disorders, for example, monitoring the therapeutic effect exhibited by the conjugate described herein, as well as its biodistribution.
  • labeling agent describes a detectable moiety or a probe.
  • exemplary labeling agents which are suitable for use in the context of the these embodiments include, but are not limited to, a fluorescent agent, a radioactive agent, a magnetic agent, a chromophore, a bioluminescent agent, a chemiluminescent agent, a phosphorescent agent and a heavy metal cluster.
  • radioactive agent describes a substance (i.e. radionuclide or radioisotope) which loses energy (decays) by emitting ionizing particles and radiation. When the substance decays, its presence can be determined by detecting the radiation emitted by it.
  • a particularly useful type of radioactive decay is positron emission.
  • Exemplary radioactive agents include 99m Tc, 18 F, 131 I and 125 L,
  • magnetic agent describes a substance which is attracted to an externally applied magnetic field. These substances are commonly used as contrast media in order to improve the visibility of internal body structures in Magnetic Resonance Imaging (MRI).
  • MRI contrast agents alter the relaxation times of tissues and body cavities where they are present, which, depending on the image weighting, can give a higher or lower signal.
  • chromophore describes a chemical moiety that, when attached to another molecule, renders the latter colored and thus visible when various spectrophotometric measurements are applied.
  • bioluminescent agent describes a substance which emits light by a biochemical process
  • chemiluminescent agent describes a substance which emits light as the result of a chemical reaction.
  • fluorescent agent refers to a compound that emits light at a specific wavelength during exposure to radiation from an external source.
  • phosphorescent agent refers to a compound emitting light without appreciable heat or external excitation as by slow oxidation of phosphorous.
  • a heavy metal cluster can be for example a cluster of gold atoms used, for example, for labeling in electron microscopy techniques.
  • the labeling agent is Fluorescein isothiocyanate.
  • a fluorescent agent being Fluorescein isothiocyanate (FITC) has been conjugated to HPMA copolymeric backbone having TNP-470 and alendronate attached thereto (HPMA copolymer-ALN- TNP-470-FITC; see. Figure 1).
  • the fluorescent agent was utilized for assessing the in vivo biodistribution of the conjugate (see, Figure 7G) as well as to study the mechanism by which the conjugate internalize into endothelial and human osteosarcoma cells (see, Figure 4).
  • These fluorescence studies showed that the conjugate is mainly distributed to bone tissue and that the mechanism by which the conjugate is internalized is through a lysosomotropic pathway of cellular uptake via clathrin-coated vesicles.
  • the conjugate has the following structure:
  • x is an integer having a value such that x/(x+y+w+z) multiplied by 100 is in the range of from 0.01 to 99.9, as described herein;
  • y is an integer having a value such that y/(x+y+w+z) multiplied by 100 is in the range of from 0.01 to 99.9, as described herein;
  • w is an integer having a value such that w/(x+y+w+z) multiplied by 100 is in the range of from 2.1 to 99, as described herein; and
  • z is an integer having a value such that z/(x+y+w+z) multiplied by 100 is in the range of from 0.01 to 99.9.
  • z is an integer having a value such that z/(x+y+w+z) multiplied by 100 is in the range of from 0.01 to 10, and depends on the labeling agent utilized and the monitoring technology.
  • conjugates described herein were successfully prepared by devising and successfully practicing novel processes for their preparation. Such processes, in addition to allowing obtaining a conjugate with a high load of alendronate, further allow for obtaining conjugates having a low polydispersity index (PDI) and small mean size distribution.
  • PDI polydispersity index
  • the conjugate described herein has a polydispersity index ranging from 1 to 1.4.
  • polydispersity index is a measure of the distribution of molecular mass in a given polymer sample.
  • PDI is a value calculated as the weight average molecular weight divided by the number average molecular weight (M w /M n ). It indicates the distribution of individual molecular masses in a batch of polymers.
  • PDI has a value always greater than 1, but as the polymer chains approach uniform chain length, the PDI approaches unity (1).
  • Polymer-based nanocarriers similar to HPMA copolymer often exhibit inherent structural heterogeneity of the polymers, reflected in a high PDI value, typically higher than 1.4, and even higher than 1.6.
  • Homogenous size distribution of polymer conjugates may contribute to a more defined biodistribution.
  • the PDI value may be 1.39, 1.3S, 1.37, 1.36, 1.35, 1.34, 1.33, 1.32, 1.31, 1.3, 1.29, 1.28, 1.27, 1.26, 1.25, 1.24, 1.26, 1.25, 1.24, 1.23, 1.22, 1.21, 1.2, 1.19, 1.18, 1.17, 1.16, 1.15, 1.14, 1.13, 1.12, 1.11, 1.1, 1.09, 1.08, 1.07, 1.06, 1.05, 1.04, 1.03, 1.02, and 1.01.
  • the conjugate described herein has a mean size distribution lower than 150 nm.
  • the mean size distribution may be lower than 149 nm, 148 nm, 147 nm, 146 nm, 145 nm, 143 nm, 142 nm, 141 nm, 140 nm, 139 nm, 138 nm, 137 nm, 136 nm, 135 nm, 134 nm, 133 nm, 132 nm, 131 nm, 130 nm, 129 nm, 128 nm, 127 nm, 126 nm, 125 nm, 124 nm, 123 nm, 122 nm, 121 nm, 120 nm, 119 nm, 118 nm, 117 nm, 116 nm, 115 nm, 114 nm, 113 nm, 112 nm, 111 nm, 110 nm, 109 nm, 108 nm, 107
  • Weight average molar mass (M w ) was evaluated for the conjugates synthesized from their SEC profiles and the Polydispersity index (PDI) was calculated according to the formula M w /M n . While reducing the present invention to practice, the present inventors have designed and successfully practiced a novel process for preparing a HPMA co-polymer having attached thereto alendronate, TNP-470 and optionally a labeling agent (e.g., a fluorescent agent), whereby the load of the alendronate is greater than in currently known methodologies for attaching bisphosphonates, and optionally other targeting moieties, to polymers.
  • a labeling agent e.g., a fluorescent agent
  • monomeric units of the polymeric backbone (HPMA) to which alendronate is attached are first prepared and then co-polymerized with HPMA monomeric, oligomeric and/or polymeric units.
  • the alendronate-containing monomeric units are polymerized and then co-polymerized with other monomeric, oligomeric or polymeric units of the monomer.
  • co-polymerization is effected upon converting at least a pre- determined portion of the HPMA monomers, oligomers or polymers to such that terminate with a reactive group that is capable of reacting with TNP-470 (e.g., by means of adding a spacer that terminates with the desired reactive group), thereby functionalizing the HPMA monomers.
  • co-polymerization is effected upon converting at least a pre-determined portion of the HPMA monomers, oligomers or polymers to such that terminate with a reactive group that is capable of reacting with a labeling agent (e.g., by means of adding a spacer that terminates with the desired reactive group).
  • co-polymerization is effected upon converting at least a pre-determined portion of the HPMA monomers, oligomers or polymers to such that include a linker, as described herein, which optionally terminates with a reactive group that is capable of reacting with TNP-470 (e.g., by means of adding a spacer that terminates with the desired reactive group).
  • a process of synthesizing the conjugates described herein comprising: (a) coupling alendronate to N-(2-hydroxypropyl)methacrylamide monomeric units, to thereby obtain alendronate-containing methacrylamide monomeric units;
  • the process further comprises, optionally prior to the co- polymerizing in (b), coupling a labeling agent to N-(2-hydroxypropyl)methacrylamide monomeric units, to thereby obtain labeling agent-containing methacrylamide monomeric units; and (b) further comprises copolymerizing the labeling agent- containing methacrylamide monomeric units together with alendronate-containing methacrylamide monomeric units and N-(2-hydroxypropyl)methacrylamide-derived monomelic units terminating with a first reactive group, to thereby obtain an alendronate-containing copolymer having the reactive group, and labeling agent, the reactive group being capable of coupling TNP-470.
  • the co-polymerization in (b) comprises co-polymerizing the alendronate-containing monomeric units with the N-(2-hydroxypropyl)methacrylamide monomeric units and/or the ((N-(2-hydroxypropyl)methacrylamide) oligomeric or polymeric units and the N-(2-hydroxypropyl)methacrylamide-derived monomeric units terminating with a first reactive group, and further with N-(2- hydroxypropyl)methacrylamide-derived monomeric units terminating with a second reactive group, wherein the second reactive group is being capable of coupling the labeling agent.
  • the process further comprises coupling the labeling agent to the co-polymer, via the second reactive group.
  • a coupling can be effected prior to, concomitant with, or subsequent to coupling the TNP-470.
  • the copolymerization of the alendronate-containing HPMA monomers and the other, functionalized or non-functionalized HPMA monomers can be effected by any polymerization method known in the art, using suitable polymerization initiators and optionally chain transfer agents. Such suitable polymerization initiators and chain transfer agents can be readily identified by a person skilled in the art.
  • step (b) in the process described hereinabove was performed via two methodologies: (1) the "classical" thermopolymerization methodology using, as an example, 4,4'-azobis(4-cyanovaleric acid) as a polymerization initiator; and (2) the "reversible addition-fragmentation chain transfer” (RAFT) polymerization technique, using, as an example, 2,2'-Azobis[2-(2- imidazolin-2-yl)propane]dihydrochloride as a polymerization initiator and 5,S"-bis( ⁇ , ⁇ '- dimethyl- ⁇ "-acetic acid) trithiocarbonate as a chain transfer agent (TTC).
  • RAFT reversible addition-fragmentation chain transfer
  • RAFT reversible addition-fragmentation chain transfer
  • thiocarbonylthio compounds such as dithioesters, dithiocarbamates, trithiocarbonates, and xanthates in order to mediate the polymerization via a reversible chain-transfer process. This allows access to polymers with low polydispersity and high functionality.
  • the co-polymerizing is performed via the Reversible addition-fragmentation chain transfer (RAFT) technique.
  • RAFT Reversible addition-fragmentation chain transfer
  • the process is performed such that the conjugate has a polydispersity index ranging from 1 to 1.4.
  • the process is performed such that the conjugate has a mean size distribution lower than 150 nm.
  • the TNP-470 or alendronate can be attached to the monomeric units that form the polymeric backbone, or to the backbone units of the copolymer, by means of a functional group that is already present in the native molecule and/or the monomeric units of the polymer, or otherwise can be introduced or generated by well- known procedures in synthetic organic chemistry without altering the activity of the agent.
  • a terminal carboxylic group can be generated within a monomeric HPMA, in order to form an amide with the amine functional group of alendronate.
  • a carboxylic functional group can be generated, for example, by oxidizing the hydroxy group of the HPMA.
  • a carboxylic functional group (reactive group) is generated by attaching to an HPMA monomer a spacer or a linker that terminates with a carboxylic group.
  • an alkylhalide can be generated within the HPMA polymeric backbone or within HPMA monomers, in order to readily couple TNP-470. Such an alkylhalide can be generated by means of a spacer and/or linker, as described herein.
  • HPMA monomeric, oligomeric and polymeric units that have been modified so as to generate a reactive group are therefore referred to herein as HPMA-derived units or as methacrylamide units terminating by a reactive group.
  • the process further comprises introduction of a linker to at least some of the HPMA monomeric, oligomeric or polymeric units participating in the co-polymerization.
  • introducing the linker is performed subsequent to the co- polymerization.
  • the process further comprises introduction of a spacer to at least some of the HPMA monomeric, oligomeric or polymeric units participating in the co-polymerization.
  • introducing the spacer is performed subsequent to the co- polymerization.
  • a plurality of functionalized HPMA monomeric units is first prepared.
  • the functionalized HPMA monomers include: HPMA monomers that include a spacer and/or a linker for attaching alendronate; HPMA monomers that include a spacer and/or a linker for attaching TNP-470; and optionally HPMA monomers that include a spacer for attaching a labeling agent. These functionalized HPMA monomeric units are referred to herein as HPMA-derived units.
  • alendronate-containing HPMA-derived monomers are prepared, and are co-polymerized with the other functionalized HPMA monomers, optionally in the presence of non-modified HPMA monomers (which form "free" backbone units upon co-polymerization).
  • alendronate-containing methacrylamide monomeric are prepared by first preparing JV-methacryloylglycylglycylprolylnorleucine units, and thereafter conjugating thereto the alendronate, so as to obtain N- methacryloylglycylglycylprolylnorleucyl-alendronate monomeric units.
  • HPMA-derived methacrylamide units that terminate with a first reactive group include JV-methacryloylglycylglycylprolylnorleucine units.
  • HPMA-derived methacrylamide units that terminate with a second reactive group include iV-methacryloylglycylglycyl units.
  • Co-polymerization is effected as described hereinabove. Then, TNP-470 is coupled to the formed co-polymer.
  • the order of steps can be modified, as long as alendronate is attached to monomeric HPMA units, prior to co-polymerization of such units, in order to assure a high and controllable load of alendronate.
  • the present inventors have utilized some of the methodologies described herein for introducing another targeting moiety into a polymer conjugate that further comprises a therapeutically active agent such as an anti-angiogenesis agent.
  • a therapeutically active agent such as an anti-angiogenesis agent.
  • the present inventors have further designed and successfully prepared and practiced novel conjugates of a polymer (e.g., a N-(2-hydroxypropyl)methacrylamide (HPMA) copolymer), an anti-angiogenesis agent (e.g., TNP-470) and a bone targeting agent being an oilgoaspartate (e.g. D-Asps).
  • HPMA N-(2-hydroxypropyl)methacrylamide
  • HPMA copolymer having TNP-470 and D-Asps attached thereto has been prepared (HPMA copolymer-D- Asp 8 -TNP-470; see, Figure S).
  • the anti-angiogenesis activity of the conjugate has been demonstrated by the ability to inhibit the proliferation of HUVEC by the conjugate (see, Figure 10) and inhibition of vascular endothelial growth factor (VEGF)-induced HUVEC migration (Figure 11).
  • VEGF vascular endothelial growth factor
  • a polymeric conjugate comprising a polymeric backbone having attached thereto an anti-angiogenesis agent and a bone targeting moiety, the bone targeting moiety being an oligopeptide of aspartic acid which comprises from 2 to 100 aspartic acid residues.
  • anti-angiogenesis agent is as defined hereinabove.
  • bone targeting moiety describes a compound having the capability of preferentially accumulating in hard tissues (i.e. bone tissues) rather than any other organ or tissue, after administration in vivo.
  • Oligopeptides of aspartic acid such as D-aspartate octapeptide (D-Asps) have been known to accumulate in bone. These oligopeptides bind to Hydroxyapitate (HA), the major constituent of the bone, thereby being suitable for serving as a bone targeting moiety.
  • HA Hydroxyapitate
  • oligopeptide of aspartic acid described herein comprises from 2 to 100 aspartic acid residues. Therefore, such oligopeptides may comprise 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100 etc. aspartic acid residues.
  • the oligopeptide of aspartic acid comprises from 2 to 20 aspartic acid residues. In some embodiments the oligopeptide of aspartic acid comprises 8 aspartic acid residues (Asps). In some embodiments the oligopeptide of aspartic acid consists of 8 aspartic acid residues (Asps).
  • the oligopeptide can further include other amino acid residues, as long as it includes one or more amino acid sequences that consist of two or more aspartic acid residues. In some embodiments, such amino acid sequences consist of 2 to 20 aspartic acid residues or of 8 aspartic acid residues.
  • the aspartic acid can be D-aspartic acid and/or L-aspartic acid. In some embodiments, the aspartic acid is D-aspartic acid.
  • the anti-angiogenesis agent is TNP-470.
  • anti-angiogenesis agents useful in the context of these embodiments of the invention include, but are not limited to, paclitaxel, 2-methoxyestradiol, prinomastat, batimastat, BAY 12-9566, carboxyamidotriazole, CC-1088, dextromethorphan acetic acid, dimethylxanthenone acetic acid, endostatin, IM-862, marimastat, a matrix metalloproteinase, penicillamine, PTK787/ZK 222584, RPI.4610, squalamine lactate, SU5416, thalidomide, combretastatin, tamoxifen, COL-3, neovastat, BMS-275291, SU6668, anti-VEGF antibody, Medi-522 (Vitaxin II), CAI, Interleukin-12, IM862, Amilloride, Angiostatin®Protein, Angiostatin Kl-3, Angiostatin Kl-5
  • Avastin, Erbitux, Vectibix, Herceptin); small molecule tyrosine kinase inhibitors of multiple proangiogenic growth factor receptors e.g. Tarceva, Nexavar, Sutent, Iressa
  • inhibitors of mTOR mimmalian target of rapamycin
  • MMP matrix metalloproteinases
  • COL3, Marimastat, Batimastat e.g. COL3, Marimastat, Batimastat
  • EMD121974 e.g. COL3, Marimastat, Batimastat
  • Vitaxin Squalamin
  • PDGFR inhibitors e.g., Gleevec
  • NM3 and 2-ME2 NM3 and 2-ME2.
  • the anti-angiogenesis agent is selected from the group consisting of Paclitaxel, monoclonal antibodies directed against specific proangiogenic factors and/or their receptors (e.g. Avastin, Erbitux, Vectibix, Herceptin); small molecule tyrosine kinase inhibitors of multiple proangiogenic growth factor receptors (e.g. Tarceva, Nexavar, Sutent, Iressa); inhibitors of mTOR (mammalian target of rapamycin) (e.g. Torisel); interferon alpha, beta and gamma; IL-12; matrix metalloproteinases (MMP) inhibitors (e.g.
  • MMP matrix metalloproteinases
  • COL3, Marimastat, Batimastat EMD121974 (Cilengitide); Vitaxin; Squalamin; COX-2 inhibitors; PDGFR inhibitors (e.g., Gleevec); NM3; 2-ME2 and Bisphosphonates (e.g., Zoledronate).
  • PDGFR inhibitors e.g., Gleevec
  • NM3 e.g., 2-ME2
  • Bisphosphonates e.g., Zoledronate
  • COX-2 inhibitor refers to a non-steroidal drug that relatively inhibits the enzyme COX-2 in preference to COX-I.
  • Preferred examples of COX-2 inhibitors include, but are no limited to, celecoxib, parecoxib, rofecoxib, valdecoxib, meloxicam, and etoricoxib.
  • the polymeric conjugates described herein are composed of a polymeric backbone, formed from a plurality of backbone units that are covalently linked to one another, wherein at least a portion of this plurality of backbone units has an anti-angiogenesis agent, as described herein, attached thereto, and at least another portion of the plurality of backbone units has the bone targeting moiety (the oligoaspartate, as described herein), attached thereto.
  • Those backbone units that have the anti-angiogenesis agent attached thereto and those backbone units that have the oligoaspartate attached thereto can be randomly dispersed within the polymeric backbone.
  • the polymeric backbone can further include non-functionalized backbone units, as discussed hereinbelow, to which none of the anti-angiogenesis agent and the oligoaspartate are attached.
  • the polymeric backbone of the conjugates described constitutes polymers (or co-polymers) to which the anti-angiogenesis agent and the bone targeting moiety are attached.
  • Polymers which are suitable for use in the context of the present embodiments are preferably biocompatible, non-immunogenic and non-toxic.
  • the polymers serve as carriers that enable specific delivery into tumor tissue, possible due to the EPR effect described hereinabove.
  • polymer describes an organic substance composed of a plurality of repeating structural units (backbone units) covalently connected to one another.
  • polymer as used herein encompasses organic and inorganic polymers and further encompasses one or more of a homopolymer, a copolymer or a mixture thereof (a blend).
  • homopolymer as used herein describes a polymer that is made up of one type of monomeric units and hence is composed of homogenic backbone units.
  • copolymer as used herein describes a polymer that is made up of more than one type of monomeric units and hence is composed of heterogenic backbone units. The heterogenic backbone units can differ from one another by the pendant groups thereof.
  • the polymer is comprised of backbone units formed by polymerizing the corresponding monomeric units whereby the anti-angiogenesis agent and the bone targeting moiety are attached to at least a portion of the backbone units. Some or all of these backbone units are typically functionalized prior to conjugation so as to have a reactive group for attaching the anti-angiogenesis agent and the bone targeting moiety. Those backbone units that are not functionalized and/or do not participate in the conjugations of the anti-angiogenesis agent and bone targeting moiety are referred to herein as "free" backbone units.
  • the polymer may be a biostable polymer, a biodegradable polymer or a combination thereof.
  • biostable describes a compound or a polymer that remains intact under physiological conditions (e.g., is not degraded in vivo).
  • biodegradable describes a substance which can decompose under physiological and/or environmental conditions into breakdown products.
  • physiological and/or environmental conditions include, for example, hydrolysis
  • biodegradable as used in the context of embodiments of the present invention, also encompasses the term “bioresorbable”, which describes a substance that decomposes under physiological conditions to break down products that undergo bioresorption into the host-organism, namely, become metabolites of the biochemical systems of the host-organism.
  • the polymers can be water-soluble or water-insoluble. In some embodiments, the polymers are water soluble at room temperature.
  • the polymers can further be charged polymers or non-charged polymers.
  • Charged polymers can be cationic polymers, having positively charged groups and a positive net charge at a physiological pH; or anionic polymers, having negatively charged groups and a negative net charge at a physiological pH.
  • Non-charged polymers can have positively charged and negatively charged group with a neutral net charge at physiological pH, or can be non-charged.
  • the polymer has an average molecular weight in the range of 100 Da to 800 kDa. In some embodiments, the polymer has an average molecular weight lower than 60 kDa. In some embodiments, the polymer's average molecular weight ranges from 10 kDa to 40 kDa. Polymeric substances that have a molecular weight higher than 10 kDa typically exhibit an EPR effect, as described herein, while polymeric substances that have a molecular weight of 100 kDa and higher have relatively long half-lives in plasma and an inefficient renal clearance. Accordingly, a molecular weight of a polymeric conjugate can be determined while considering the half-life in plasma, the renal clearance, and the accumulation in the tumor of the conjugate.
  • the molecular weight of the polymer can be controlled, at least to some extent, by the degree of polymerization (or co-polymerization).
  • the polymer used in the context of these embodiments of the invention can be a synthetic polymer or a naturally-occurring polymer. In some embodiments, the polymer is a synthetic polymer.
  • the polymeric backbone of the polymer described herein may be derived from, for example, polyacrylates, polyvinyls, polyamides, polyurethanes, polyimines, polysaccharides, polypeptides, polycarboxylates, and mixtures thereof.
  • Exemplary polymers which are suitable for use in the context of the present embodiments include, but are not limited to the group consisting of dextran, a water soluble polyamino acid, a polyethylenglycol (PEG), a polyglutamic acid (PGA), a polylactic acid (PLA), a polylactic-co-glycolic acid, (PLGA), a poly(D,L-lactide-c ⁇ glycolide) (PLA/PLGA), a poly(hydroxyalkylmethacrylamide), a polyglycerol, a polyamidoamine (PAMAM), and a polyethylenimine (PEI).
  • dextran a water soluble polyamino acid
  • PEG polyethylenglycol
  • PGA polyglutamic acid
  • PLA polylactic acid
  • PLA polylactic-co-glycolic acid
  • PLA/PLGA poly(D,L-lactide-c ⁇ glycolide)
  • PAMAM polyamidoamine
  • These polymers can be of any molecular weight, as described herein.
  • the polymeric backbone is derived from a poly(hydroxyalkylmethacrylamide) or a copolymer thereof.
  • a polymeric backbone comprises methacrylamide backbone units having attached thereto either 2- hydroxypropyl groups or such 2-hydroxypropyl groups that have been modified by attaching thereto (directly or indirectly) the moieties described herein (the oligoaspartate and the anti-angiogenesis agent).
  • polymers as discussed herein describe those polymers that are formed from homogenic or heterogenic, non-functionalized monomeric units, and that the polymeric backbone constituting the polymeric conjugate corresponds to such polymers by being comprised of the same monomeric units, while some of these monomeric units are functionalized, as described herein.
  • the polymeric backbone of the polymeric conjugate is similar to that of the polymers described herein, and differs from the polymers by having the above-described agents attached to some of the backbone units therein.
  • the bone targeting moiety and the anti-angiogenesis agent can each be linked to the respective portion of the backbone units in the polymeric backbone directly, or indirectly, through a linker moiety (also referred to herein as a linker, a linker group or a linking group), whereby, in some embodiments, the direct/indirect linkage is designed as being cleavable at conditions characterizing the desired bodily site (e.g., by certain enzymes or pH), as detailed hereinbelow.
  • a linker moiety also referred to herein as a linker, a linker group or a linking group
  • At least one of the anti- angiogenesis agent and the bone targeting moiety is attached to the polymer via a linker.
  • each of the anti-angiogenesis agent and the bone targeting moiety is attached to the polymer via a linker.
  • the linker linking the anti- angiogenesis agent to the polymer and the linker linking the bone targeting moiety to the polymer may be the same or different.
  • the linker described herein refers to a chemical moiety that serves to couple the anti-angiogenesis agent and/or the bone targeting moiety to the polymer while not adversely affecting either the targeting function of the bone targeting moiety or the therapeutic effect of the anti-angiogenesis agent.
  • each of the anti-angiogenesis agent and the bone targeting moiety is attached to the polymer via a linker.
  • the linker linking the anti-angiogenesis agent and the linker linking the bone targeting moiety may be the same or different.
  • only the anti-angiogenesis agent is linked to the polymer via a biodegradable linker, thereby being released from the polymer at the desired bodily site.
  • a HPMA copolymer of TNP-470 and D-Asp 8 has been synthesized using the biodegradable linker -[GIy-GIy- Pro-Nle]- in order to link TNP-470 to the polymer.
  • an exemplary linker is a Cathepsin K cleavable linker.
  • the linker is an enzymatically-cleavable linker.
  • the enzymatically-cleavable linker is cleaved by an enzyme which is expressed in tumor tissues.
  • the enzymatically-cleavable linker is cleaved by an enzyme which is overexpressed in tumor tissues.
  • Exemplary enzymes which are suitable for use in the context of the present embodiments include, but are not limited to Cathepsin B, Cathepsin K, Cathepsin D, Cathepsin H, Cathepsin L, legumain, MMP-2 and MMP-9.
  • Cathepsin K is expressed predominantly in osteoclasts. Therefore, in some embodiments the enzymatically-cleavable linker is cleaved by Cathepsin K.
  • the biodegradable linker comprises an oligopeptide having from 2 to 10 amino acid residues.
  • a Cathepsin K cleavable linker being -[Gly-Gly-Pro-
  • the linker comprises - [Gly-Gly-Pro-Nle]-.
  • the bone targeting moiety described in the context of these embodiments of the invention is attached to the polymeric backbone via a biostable linker.
  • the bone targeting moieties attached to the polymeric backbone via an enzymatically cleavable linker for example, a Cathepsin K cleavable linker as described hereinabove.
  • the conjugate described herein comprise an additional spacer moiety which enables a more efficient and simpler attachment of the anti-angiogenesis agent and/or the bone targeting moiety to the polymeric backbone.
  • the spacer may be further utilized in order to attach a labeling agent to the conjugate. Such a spacer has been described extensively hereinabove.
  • spacers comprising a -[NH-(CH 2 ) 2 -NH]- group and a 1-aminohaxanoyl have been used, intradispersly, in order to conjugate TNP-470 through a -[Gly-Gly-Pro-Nle]- Cathepsin K cleavable linker to the HPMA copolymer (see, Figure 8).
  • Spacers derived from 1- aminohexaonoic acid were used intradispersly in order to attach D-Asps and a labeling agent being FITC to the polymer (see, Figure 8).
  • the conjugate may further comprise a labeling agent, as defined herein.
  • a labeling agent is described elaborately hereinabove.
  • the labeling agent is attached to the conjugate via a spacer, as described herein.
  • FITC Fluorescein isothiocyanate
  • FITC Fluorescein isothiocyanate
  • HPMA copolymer-D-Asps- TNP-470 via the spacer used to couple the D-Asps (see, Figure 8).
  • the degree of loading of the anti-angiogenesis agent and the bone targeting moiety may be expressed as mol %, as defined herein.
  • a 1 mol % load of a bone targeting moiety describes a polymeric conjugate composed of 100 backbone units, whereby 1 backbone unit has a targeting moiety attached thereto and the other 99 backbone units are either free or have other agents attached thereto.
  • the optimal degree of loading of the anti-angiogenesis agent and bone targeting moiety for a given conjugate and a given use is determined empirically based on the desired properties of the conjugate (e.g., water solubility, therapeutic efficacy, pharmacokinetic properties, toxicity and dosage requirements), and optionally on the amount of the conjugated moiety that can be attached to a polymeric backbone in a synthetic pathway of choice.
  • the % loading can be measured by methods well known by those skilled in the art, some of which are described hereinbelow under the Materials and Methods of the Examples section that follows.
  • the loading of the anti-angiogenesis agent in the polymer is greater than 1 mol %.
  • the loading of the anti-angiogenesis agent in the conjugate ranges from 1 mol % to 99 mol %, from 1 mol % to 50 mol %, from 1 mol % to 20 mol %, from 1 mol % to 10 mol %, or from 1 mol % to 5 mol %. In some embodiments the loading of the bone targeting moiety in the polymer is greater than 1 mol %.
  • the loading of the bone targeting moiety in the conjugate ranges from 1 mol % to 99 mol %, from 1 mol % to 50 mol %, from 1 mol % to 20 mol %, from 1 mol % to 10 mol %, or from 1 mol % to 5 mol %.
  • the number of backbone units within the polymeric backbone that have an anti- angiogenesis agent conjugated thereto is defined herein as "a”
  • the number of backbone units within the polymeric backbone that have a bone targeting moiety conjugated thereto is herein defined as “b”
  • the number of free backbone units in the polymeric backbone (which are not bound to an additional moiety) is herein defined as "d”.
  • the conjugate described herein can be represented by the general formula I:
  • a is an integer having a value such that x/(x+y+w) multiplied by 100 is in the range of from 0.01 to 99.9
  • b is an integer having a value such that y/(x+y+w) multiplied by 100 is in the range of from 0.01 to 99.9
  • d is an integer having a value such that w/(x+y+w) multiplied by 100 is in the range of from 0.01 to 99.9;
  • a 1 , A 2 and A 3 are each backbone units covalently linked to one another and forming the polymeric backbone, wherein:
  • B is the anti-angiogenesis agent as defined hereinabove
  • D is the bone targeting moiety as defined hereinabove
  • each of the L 1 and L 2 is independently a linker as defined hereinabove; such that [A 2 -Li-B] is a backbone unit having attached thereto the anti- angiogenesis agent
  • [A 3 -L 2 -D] is a backbone unit having attached thereto the bone targeting moiety; wherein each of the [A 1 ], the [A 2 -L 1 -B] and the [A 3 -L 2 -D] is either a terminal backbone unit being linked to one of the [A 1 ], the [A 2 -L 1 -B] and the [A 3 -L 2 -D], or is linked to at least two of the [Ai], the [A 2 -L 1 -B] and the [A 3 -L 2 -D] and the Ai, A 2 and/or A 3 are linked to one another to thereby form the polymeric backbone.
  • a 1 is a hydroxypropylmethacrylamide unit; and A 2 and A 3 is a methacrylamide unit, as discussed hereinabove.
  • the conjugate described herein can be represented by the general formula Ha:
  • conjugate has the following structure:
  • a and b are each independently an integer having a value such that a/(a+b+d) multiplied by 100 and/or b/(a+b+d) xlOO are in the range of from 0.01 to 15; and d is an integer having a value such that d/(a+b+d) multiplied by 100 is in the range of from 70 to 99.9. It would be appreciated that a, b and d can be controlled as desired by selecting the mol ratio of the respective monomeric units used for forming the polymeric conjugate.
  • oligomeric units of the polymer or simply “oligomeric units”, as used herein throughout, describes a polymer comprised of 2-50 backbone units.
  • the polymeric backbone of the conjugate described herein may be constructed by copolymerizing the functionalized monomeric units, as described hereinabove, together with non-functionalized monomeric or oligomeric units that compose the backbone.
  • each of the functionalized monomeric units can first be polymerized, so as to form a functionalized oligomer bearing a plurality of the first reactive groups and a functionalized oligomer bearing a plurality of the second reactive groups, and these oligomers can be co-polymerized with each other and with the non- functionalized oligomeric or monomeric units.
  • only one of the functionalized monomeric units is polymerized so as to form a functionalized oligomer, which is then co-polymerized with the other functionalized monomeric units and non-functionalized monomeric or oligomeric units.
  • the term "functionalized” describes a monomer or an oligomer that terminates with one or more reactive groups.
  • a "reactive group” describes a chemical group that is capable of reacting with another group so as to form a chemical bond, typically a covalent bond.
  • a covalent bond typically a covalent bond.
  • an ionic or coordinative bond is formed.
  • a reactive group is termed as such if being chemically compatible with a reactive group of an agent or moiety that should be desirably attached thereto.
  • a carboxylic group is a reactive group suitable for conjugating an agent or a moiety that terminates with an amine group, and vice versa.
  • a reactive group can be inherently present in the monomeric units, oligomeric units and/or bone targeting moiety and the anti-angiogenesis agent, or be generated therewithin by terms of chemical modifications of the chemical groups thereon or by means of attaching to these chemical groups a spacer or a linker that terminates with the desired reactive group, as described herein.
  • Co-polymerizing the monomers or oligomers described herein can be effected by any of the polymerization methods known in the art, using suitable polymerization initiators or any other catalysts known in the art.
  • the co-polymerization is performed via the reversible addition-fragmentation chain transfer (RAFT) technique, as exemplified in the Examples section that follows.
  • RAFT reversible addition-fragmentation chain transfer
  • the anti-angiogenesis agent is conjugated to the polymer prior to the conjugation of the bone targeting moiety.
  • the bone targeting moiety is coupled to the polymer prior to conjugating the anti-angiogenesis agent.
  • Each of the first and the second reactive groups can be protected prior to the respective conjugation thereto.
  • the process further comprises deprotecting each of the reactive groups prior to the respective conjugation.
  • the monomeric units, spacers and linkers utilized for coupling the anti-angiogenesis agent and/or the bone targeting moiety to the polymer are designed so as to allow a smooth and efficient conjugation of the respective moiety and an optimal performance of the obtained conjugate, as discussed elaborately hereinabove.
  • the process is further effected by preparing the monomeric units or oligomeric units that comprise the first and second reactive groups.
  • monomeric units having attached thereto a spacer terminating with a protected first reactive group are prepared.
  • Exemplary such monomeric units are methacrylamide units (derived from HPMA, as defined herein) having attached thereto a protected Gly-Gly group.
  • the ratio between the above-described monomeric units and non-functionalized monomeric or oligomeric units that form a part of the formed polymer determines, at least in part, the mol ratio of the respective bone targeting moiety and anti-angiogenesis moiety in the formed conjugate.
  • Co-polymerizing the monomeric and/or oligomeric units bearing the reactive groups results in a functionalized polymer, bearing the first and second reactive groups (optionally protected with respective protecting groups).
  • the bone targeting moiety and/or the anti-angiogenesis moiety are modified prior to being conjugated to the functionalized polymer, so as to include reactive groups that are compatible with the first and second reactive groups, respectively, of the functionalized polymer.
  • a modification can be effected by means of attaching a spacer and/or a linker to the bone targeting moiety and/or the anti-angiogenesis agent prior to the conjugation thereof to the functionalized polymer.
  • the process is further effected by preparing such modified bone targeting moiety and/or anti-angiogenesis agent.
  • linkers and/or spacers interposed between the polymeric backbone and the moieties conjugated thereto are designed so as to exhibit the properties described elaborately hereinabove with respect thereto.
  • the spacer may be varied in length and in composition depending on steric consideration and may be used to space the angiogenesis agent and/or bone targeting moiety form the polymer, thereby enabling easier synthesis of the conjugate and/or improved performance of the formed conjugate, as detailed hereinabove.
  • the process further comprises attaching a labeling agent, as defined herein, to the formed conjugate.
  • the labeling agent can be attached to either of functionalized monomeric units, prior to co-polymerization or to the formed copolymer.
  • the labeling agent is attached to the co-polymer concomitantly with the bone targeting moiety. Alternatively, it is attached prior to or subsequent to attaching the bone targeting moiety and/or the anti-angiogenesis agent.
  • the process comprises co-polymerizing, along with the functionalized and non-functionalized monomeric or oligomeric units described herein, monomeric units terminating with a third reactive group, the third reactive group being for conjugating thereto a labeling agent or any other additional moiety, as described herein.
  • each of the conjugates described in any of the embodiments of the invention may further include an additional moiety conjugated thereto.
  • Such an additional moiety can be conjugated either to monomeric units within and throughout the polymeric backbone, or be attached at one or both ends of the polymeric backbone.
  • Such an additional moiety can be a labeling agent, as described herein, or an additional targeting moiety or an additional therapeutically active agent, which may improve the performance of the formed conjugate.
  • Such an additional moiety can further be a moiety that improves the solubility, bioavailability, and/or any other desired feature of the formed conjugate.
  • the conjugates described hereinabove may be prepared, administered or otherwise utilized in any of the aspects of embodiments of the invention, either as is, or as a pharmaceutically acceptable salt, enantiomers, diastereomers, solvates, hydrates or a prodrug thereof.
  • pharmaceutically acceptable salt refers to a charged species of the parent compound and its counter ion, which is typically used to modify the solubility characteristics of the parent compound and/or to reduce any significant irritation to an organism by the parent compound, while not abrogating the biological activity and properties of the administered compound.
  • the neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
  • pharmaceutically acceptable salts is meant to encompass salts of the moieties and/or conjugates which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral (i.e., non-ionized) form of such conjugates with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such conjugates with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include, but are not limited to, those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et ah, "Pharmaceutical Salts", Journal of Pharmaceutical Science, 1977, 66, 1-19).
  • Certain specific conjugates of the present invention contain both basic and acidic functionalities that allow the conjugates to be converted into either base or acid addition salts.
  • the neutral forms of the conjugates are preferably regenerated by contacting the salt with a base or acid and isolating the parent conjugate in a conventional manner.
  • the parent form of the conjugate differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the conjugate for the purposes of the present invention.
  • a pharmaceutically acceptable salt of alendronate is utilized.
  • An exemplary such salt is sodium alendronate.
  • An alendronate-containing conjugate can therefore comprise a sodium salt of alendronate.
  • a pharmaceutically acceptable salt of aspartate is utilized.
  • prodrug refers to an agent, which is converted into the active compound (the active parent drug) in vivo.
  • Prodrugs are typically useful for facilitating the administration of the parent drug.
  • the prodrug may also have improved solubility as compared with the parent drug in pharmaceutical compositions.
  • Prodrugs are also often used to achieve a sustained release of the active compound in vivo.
  • conjugates described herein may possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, diastereomers, geometric isomers and individual isomers are encompassed within the scope of embodiments of the invention.
  • the term "enantiomer” describes a stereoisomer of a compound that is superposable with respect to its counterpart only by a complete inversion/reflection (mirror image) of each other. Enantiomers are said to have "handedness” since they refer to each other like the right and left hand. Enantiomers have identical chemical and physical properties except when present in an environment which by itself has handedness, such as all living systems.
  • conjugates described herein can exist in unsolvated forms as well as solvated forms, including hydrated forms.
  • the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention.
  • solvate refers to a complex of variable stoichiometry (e.g., di-, tri- ? tetra-, penta-, hexa-, and so on), which is formed by a solute (the conjugate described herein) and a solvent, whereby the solvent does not interfere with the biological activity of the solute.
  • Suitable solvents include, for example, ethanol, acetic acid and the like.
  • hydrate refers to a solvate, as defined hereinabove, where the solvent is water.
  • the conjugates described herein (also referred to herein throughout as polymeric conjugates) comprise a bone targeting moiety being either alendronate or an oligopeptide of aspartate, which enables the targeting of the conjugate to bone and bone related structures. Due to the anti-angiogenesis/anti- proliferative activity exhibited by the moieties attached to the polymer and the formed conjugate as a whole, each of the conjugates described herein can be beneficially used for treating bone and bone related disease and disorders.
  • any of the conjugates described herein as a medicament.
  • the medicament is for treating a bone-related disease or disorder.
  • the conjugates described herein are identified for use in the treatment of a bone related disease or disorder.
  • the term "method" refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.
  • a bone related disease or disorder describes a disease or disorder wherein bone formation, deposition, or resorption is abnormal, especially those characterized by excessive angiogenesis.
  • bone related disease or disorder also encompasses disease and disorders occurring in bodily sites other than bone which evolved from a bone related disease or disorder such as, for example, metastasis of bone cancer in another organ and diseases and disorders which evolved in other bodily sites and affect bone tissues.
  • Bone-related diseases disorders include, but are not limited to, bone cancer and bone cancer metastases, osteopenia due to bone metastases, periodontal disease, periarticular erosions in rheumatoid arthritis, Paget's disease, malignant hypercalcemia, osteolytic lesions produced by bone metastasis, bone abnormalities caused by cancer therapeutics and hyperostosis.
  • the term "treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.
  • the treatable disease is bone cancer, this term encompasses any inhibition of tumor growth or metastasis, or any attempt to inhibit, slow or abrogate tumor growth or metastasis.
  • the toxicity of the anti-angiogenesis agent is substantially reduced, due to the conjugate selectivity towards bone tissues. Consequently, besides the use of the conjugates described herein in a clinically evident disease, optionally in combination with other drugs, these conjugates may potentially be used as a long term-prophylactic for individuals who are at risk for relapse due to residual dormant cancers.
  • the use of non-toxic targeted conjugates for the treatment of asymptomatic individuals who are at risk for relapse of osteosarcoma may lead to a major paradigm shift in cancer treatment from current methods where treatment is generally not initiated until a bone related disease such as osteosarcoma becomes clinically evident.
  • subject refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.
  • the conjugates described herein inhibit angiogenesis as well as cell proliferation and therefore can be utilized for the treatment of bone related disease and disorders characterized by pathologically excessive angiogenesis wherein the inhibition of angiogenesis and/or cell proliferation is beneficial.
  • the bone related disease or disorder is associated with angiogenesis.
  • Tumor growth and metastasis are particularly dependent on the degree of angiogenesis.
  • Tumor angiogenesis is the proliferation of a network of blood vessels that penetrate into cancerous tumors in order to supply nutrients and oxygen and remove waste products, thus leading to tumor growth.
  • Tumor angiogenesis involves hormonal stimulation and activation of oncogenes, expression of angiogenic growth factors, extravasation of plasma protein, deposition of a provisional extracellular matrix (ECM), degradation of ECM, and migration, proliferation and elongation of endothelial capillaries. Inhibition of further vascular expansion has therefore been the focus of active research for cancer therapy.
  • ECM provisional extracellular matrix
  • the bone related disease or disorder is selected from the group consisting of bone cancer metastases and bone cancer.
  • cancer and “tumor” are used interchangeably herein to describe a class of diseases in which a group of cells display uncontrolled growth (division beyond the normal limits).
  • cancer encompasses malignant and benign tumors as well as disease conditions evolving from primary or secondary tumors.
  • malignant tumor describes a tumor which is not self-limited in its growth, is capable of invading into adjacent tissues, and may be capable of spreading to distant tissues (metastasizing).
  • benign tumor describes a tumor which is not-malignant (i.e. does not grow in an unlimited, aggressive manner, does not invade surrounding tissues, and does not metastasize).
  • primary tumor describes a tumor that is at the original site where it first arose.
  • secondary tumor describes a tumor that has spread from its original (primary) site of growth to another site, close to or distant from the primary site.
  • bone cancer describes tumors that arise from the tissues of the bone.
  • the term “bone cancer”, as used herein, further encompasses tumors in tissues located in proximity to bone structures and associated with bone such as cartilage, bone cavity and bone marrow.
  • the term “bone cancer” further encompasses cancer which evolved from bone cells (i.e. primary tumor), as well as cancer cells which have "breaken away”, “leaked”, or “spilled” from a primary tumor located in bone, entered the lymphatic and/or blood vessels, circulated through the lymphatic system and/or bloodstream, settled down and proliferated within normal tissues elsewhere in the body thereby creating a secondary tumor.
  • metastases originating from osteosarcoma can be frequently found in the lungs and in other organs. These lesions produce an osteoid and therefore can be targeted with bone targeting moieties, as described herein.
  • Bone cancer is found most often in the bones of the arms and legs, but it can occur in any bone.
  • Bone cancers are also known as sarcomas. There are several types of sarcomas of bone, depending upon the kind of bone tissue where the tumor developed. Exemplary types of bone cancers that are treatable according to embodiments of the invention include, but are not limited to, osteosarcoma, Ewing's sarcoma, chondrosarcoma, fibrosarcoma, malignant giant cell tumor, and chordoma.
  • Osteosarcoma is the most common type of primary bone cancer and classified as a malignant mesenchymal neoplasm in which the tumor directly produces defective osteoid (immature bone). It is a highly vascular and extremely destructive malignancy that most commonly arises in the metaphyseal ends of long bones.
  • Several strategies were proposed, such as immune-based therapy, tumor-suppressor or suicide gene therapy, or anticancer drugs that are not commonly used in osteosarcoma [Quan et al. Cancer Metastasis Rev 2006; 10: 707-713]. However, still one-third of patients die from this devastating cancer, and for those with unresectable disease there are no curative systemic therapies.
  • bone metastases describes cancer evolving form a primary tumor located in bodily site other than bone but metastasizing to the bone (i.e. a secondary tumor). Cancers that commonly metastasize, or spread, to the bones include breast cancer, lung cancer, thyroid cancer, prostate cancer, some brain cancers and cancers of the kidney.
  • prostate cancer is the most common cancer of males in industrialized countries and the second leading cause of male cancer mortality.
  • Prostate cancer predominantly metastasizes to bone, but other organ sites are affected including the lung, liver, and adrenal gland.
  • Bone metastases incidence in patients with advanced metastatic disease is approximately 70 %. Bone metastases are associated with considerable skeletal morbidity, including severe bone pain, pathologic fracture, spinal cord or nerve root compressions, and hypercalcemia of malignancy.
  • the conjugates described herein may be further utilized for monitoring bone related disease or disorders.
  • the conjugate further comprises a labeling agent, as defined herein, for easy detection of the conjugate in the body of the patient, using well known imaging techniques.
  • the detection of the conjugate as assessed by the level of labeling agent signal, can serve to detect bone cancer metastases in bodily sites other than bone.
  • a bone related disease or disorder in a subject there are provided methods of monitoring a bone related disease or disorder in a subject.
  • the method according to these embodiments of the invention is effected by administering to the subject any of the conjugates described herein, having a labeling agent attached to the polymer, as described herein, and employing an imaging technique for monitoring a distribution of the conjugate within the body or a portion thereof.
  • uses of any of the conjugates described herein, having a labeling agent as described herein, as diagnostic agents and/or in the manufacture of a diagnostic agent for monitoring a bone related disease or disorder are provided.
  • each of the conjugates described herein, which comprises a labeling agent is identified for use as a diagnostic agent, for monitoring a bone related disease or disorder.
  • Suitable imaging techniques include, but are not limited to, positron emission tomography (PET), gamma-scintigraphy, magnetic resonance imaging (MRI), functional magnetic resonance imaging (FMRI), magnetoencephalography (MEG), single photon emission computerized tomography (SPECT) computed axial tomography (CAT) scans, ultrasound, fluoroscopy and conventional X-ray imaging.
  • PET positron emission tomography
  • MRI magnetic resonance imaging
  • FMRI functional magnetic resonance imaging
  • MEG magnetoencephalography
  • SPECT single photon emission computerized tomography
  • CAT computed axial tomography
  • the appropriate imaging technique is MRI; if the labeling agent comprises radionuclides, an appropriate imaging technique is gamma-scintigraphy; if the labeling agent comprises an ultrasound agent, ultrasound is the appropriate imaging technique, etc.
  • any of the conjugates described herein can be provided to an individual either per se, or as part of a pharmaceutical composition where it is mixed with a pharmaceutically acceptable carrier.
  • a "pharmaceutical composition” refers to a preparation of one or more of the conjugates described herein (as active ingredient), or physiologically acceptable salts or prodrugs thereof, with other chemical components including but not limited to, physiologically suitable carriers, excipients, lubricants, buffering agents, antibacterial agents, bulking agents (e.g. mannitol), antioxidants (e.g., ascorbic acid or sodium bisulfite), anti-inflammatory agents, anti-viral agents, chemotherapeutic agents, anti-histamines and the like.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to a subject.
  • active ingredient refers to a compound, which is accountable for a biological effect.
  • physiologically acceptable carrier and “pharmaceutically acceptable carrier” which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • excipient refers to an inert substance added to a pharmaceutical composition to further facilitate administration of a drug. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols. Techniques for formulation and administration of drugs may be found in
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the compounds into preparations which can be used pharmaceutically.
  • Proper formulation is dependent upon the route of administration chosen.
  • the dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition (see e.g., Fingl et al., 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 p.l).
  • the pharmaceutical composition may be formulated for administration in either one or more of routes depending on whether local or systemic treatment or administration is of choice, and on the area to be treated. Administration may be done orally, by inhalation, or parenterally, for example by intravenous drip or intraperitoneal, subcutaneous, intramuscular or intravenous injection, or topically (including ophtalmically, vaginally, rectally, intranasally).
  • Formulations for topical administration may include but are not limited to lotions, ointments, gels, creams, suppositories, drops, liquids, sprays and powders.
  • Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.
  • Compositions for oral administration include powders or granules, suspensions or solutions in water or non-aqueous media, sachets, pills, caplets, capsules or tablets. Thickeners, diluents, flavorings, dispersing aids, emulsifiers or binders may be desirable.
  • Formulations for parenteral administration may include, but are not limited to, sterile solutions which may also contain buffers, diluents and other suitable additives. Slow release compositions are envisaged for treatment.
  • compositions to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.
  • the pharmaceutical composition may further comprise additional pharmaceutically active or inactive agents such as, but not limited to, an anti-bacterial agent, an antioxidant, a buffering agent, a bulking agent, a surfactant, an anti- inflammatory agent, an anti-viral agent, a chemotherapeutic agent and an anti- histamine.
  • additional pharmaceutically active or inactive agents such as, but not limited to, an anti-bacterial agent, an antioxidant, a buffering agent, a bulking agent, a surfactant, an anti- inflammatory agent, an anti-viral agent, a chemotherapeutic agent and an anti- histamine.
  • the pharmaceutical composition described herein is packaged in a packaging material and identified in print, in or on the packaging material, for use in the treatment of a bone related disease or disorder.
  • the pharmaceutical composition is packaged in a packaging material and identified in print, in or on the packaging material, for use in monitoring a bone related disease or disorder.
  • compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may, for example, comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration.
  • Such notice for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert.
  • the conjugates described herein can be utilized in combination with additional therapeutically active agents.
  • additional agents include, as non-limiting examples, chemotherapeutic agents, anti-angiogensis agents, hormones, growth factors, antibiotics, anti-microbial agents, anti-depressants, immunostimulants, and any other agent that may enhance the therapeutic effect of the conjugate and/or the well-being of the treated subject.
  • compositions, methods or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.
  • a compound or “at least one compound” may include a plurality of compounds, including mixtures thereof.
  • range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.
  • Flash chromatography silica gel Merck 60 (particle size 0.040-0.063 mm), eluent given in parentheses.
  • Cathepsin K inhibitor III was purchased from Calbiochem, Germany.
  • Phalloidin-TRITC conjugate Propidium iodide and hydroxyapatite (HA) were purchased from Sigma-Aldrich, Israel. Alendronate was purchased from Alcon Biosciences, India.
  • Ultra pure double distilled water was purchased from Biological Industries, Israel.
  • Antifade ® mounting media was from Biomeda. Alexa ® Fluor 594 human transferrin was from Molecular ProbesTM.
  • Peroxidase Block was purchased from Merck, Germany. Primary rat anti-murine
  • CD34 antibody (MEC 14.7) was purchased from Abeam, (Cambridge, MA). Rabbit anti-rat antibody, anti-rabbit horseradish peroxidase-conjugated antibody (ABC detection kit) and ImmPACTTM DAB diluent kit were purchased from Vector Laboratories, CA, USA.
  • Boyden chambers 8 ⁇ m were from Transwell-Costar Corporation.
  • Hema 3 Stain System was from Fisher Diagnostics.
  • EGM-2 medium was from Cambrex, USA and endothelial cells growth supplement (ECGS) from Zotal, Israel. All other chemical reagents, including salts and solvents, were purchased from
  • mice's body weight and tumor size were measured three times a week.
  • mCherry was subcloned from pART7-mCherry, into pQCXIP (Clontech).
  • Human embryonic kidney 293T (HEK 293T) cells were co-transfected with pQC- mCherry and the compatible packaging plasmids (pMD.G.VSVG and pGag-pol.gpt).
  • pQC-mCherry retroviral particles containing supernatant was collected.
  • MG-63-Ras human osteosarcoma cells were infected with the retroviral particles media, and 48 hours following the infection, mCherry positive cells were selected by puromycin resistance.
  • HUVEC were plated at 10,000 cells/well onto 24-well culture plates in EBM-2 supplemented with 5 % FBS and incubated for 24 hours (37 0 C; 5 % CO 2 ).
  • Medium was replaced with 2.5 % endothelial cell basal medium-2 (EBM-2) supplemented with 1
  • ECGS Endothelial cell growth supplement
  • 63-Ras cells were plated at 2500 cells/well in DMEM supplemented with 5 % FBS and incubated for 24 hours (37 0 C; 5 % CO 2 ). The medium was then replaced with DMEM supplemented with 10 % FBS. Cells were exposed to ALN, TNP-470, and HPMA copolymer-ALN-TNP-470 conjugate or with equivalent concentrations of combinations of free ALN and TNP-470 at serial dilutions. HUVEC were also incubated with or without 1 ⁇ M of cathepsin-K inhibitor III. Control cells were grown in the presence or absence of growth factors. HUVEC or Saos-2 viable cells were counted by a Zl
  • IC 50 represents the concentration of a drug that is required for 50 % inhibition in vitro.
  • the IC 3O so, 70 values of treatment with ALN, TNP-470 and their respective combinations from HUVEC proliferation assay were collected.
  • IC 30 , 50 , 70 values of TNP-470 and ALN were marked on X, Y axis respectively and a line which represents additive effect was drawn between each inhibitory concentration (IC).
  • the content of TNP-470 was estimated from the content of NH 2 groups in the HPMA copolymer-ALN-NH 2 precursor (shown in Figure 1, middle structure), assuming that the TNP-470 binding was quantitative.
  • the content of NH 2 groups was determined by ninhydrin method using an amine containing monomer (N-(3- aminopropyl)methacrylamide) as the calibration sample (modified from Duncan, at el. J Control Release 2001;74: 135-146).
  • the mean hydrodynamic diameter of the conjugate was evaluated using a real time particle analyzer (NANOSIGHT LM20TM) containing a solid-state, single mode laser diode ( ⁇ 20 mW, 655 nm) configured to launch a finely focused beam through a 500 ⁇ l sample chamber.
  • NANOSIGHT LM20TM real time particle analyzer
  • HPMA copolymer-ALN-TNP-470 conjugate was dissolved in phosphate buffered saline (PBS) to final concentrations of 0.5, 1 and 2 mg/ml. The samples were then injected into the chamber by syringe and allowed to equilibrate to unit temperature (23 °C) for 30 seconds.
  • the particles dynamics were visualized at 30 frames per second (fps) for 60 seconds at 640 x 480 resolution by a coupled charge device (CCD) camera.
  • the paths the particles take under Brownian motion over time were analyzed using Nanoparticle Tracking Analysis (NTA) software.
  • NTA Nanoparticle Tracking Analysis
  • the diffusion coefficient and hence sphere equivalent hydrodynamic radius of each particle was separately determined and the particle size distribution profiles were generated. Each sample was measured three times in triplicates, and the results represent the mean diameter.
  • HPMA copolymer- ALN-TNP-470 conjugate In order to assess the ability of HPMA copolymer- ALN-TNP-470 conjugate to bind to bone mineral, its binding potency to hydroxyapatite (HA) was evaluated.
  • HPMA copolymer-ALN-TNP-470 conjugate was dissolved in phosphate buffered saline (PBS), pH 7.4 (1 mg/ml). The conjugate solution (500 ⁇ l) was incubated with hydroxyapatite powder (15 mg), in 500 ⁇ l PBS, pH 7.4. HPMA copolymer-Gly-Phe- Leu-Gly was used as control.
  • PBS phosphate buffered saline
  • HPMA copolymer-Gly-Phe- Leu-Gly was used as control.
  • cells were permeabilized with 0.1 % Triton-XIOO for 3 minutes and rinsed with PBS again.
  • nuclei were labeled using propidium iodide (10 ⁇ g/ml) and cover glasses were mounted by Antifade ® mounting media.
  • actin filaments were labeled using phalloidin-TRITC conjugate (50 ⁇ g/ml, 40 minutes at RT) and cover glasses were mounted by Vectashild ® DAPI containing medium.
  • HUVEC and Saos-2 cells were incubated with 10 ⁇ M FITC-HPMA copolymer ALN- TNP-470 conjugate for 6 hours. Following incubation cells were washed several times with cold PBS, starved for 45 minutes in serum free medium at 37 °C and incubated with 40 ⁇ g/ml Alexa ® Fluor 594 human transferrin for lhour at 37 °C. Cells were then fixed and mounted as described before. All slides were kept at 4 °C in dark until confocal microscopy analysis was preformed.
  • Human umbilical vain endothelial cells (HUVEC) migration assay Cell migration assays were performed using modified 8 ⁇ m Boyden chambers coated with 10 ⁇ g/ml fibronectin. HUVEC (15 x 10 4 cells/100 ⁇ l) were challenged with HPMA copolymer ALN-TNP-470 conjugate or with combinations of free ALN + free TNP-470 at equivalent concentrations and were added to the upper chamber of the transwell for 4 hours incubation. . Following incubation, cells were allowed to migrate to the underside of the chamber for 4 hours in the presence or absence of Vascular endothelial growth factor (VEGF) (20 ng/ml) in the lower chamber.
  • VEGF Vascular endothelial growth factor
  • TNP-470 subcutaneously
  • n 5 mice/group.
  • a modified Miles assay was performed as previously described [Claffey et al. 1996, Cancer Res 56: 172-181; Miles & Miles 1952, J Physiol 118:228-257]. Briefly, Evans blue dye (100 ⁇ l of a 1 % solution in 0.9 % NaCl) was injected into the retro-orbital plexus of the mice.
  • Tumor progression was monitored by caliper measurement (width x length 2 x 0.52) and by CRI Maestro non-invasive intravital imaging system. At termination, tumors were dissected, weighed and analyzed. Data is expressed as mean ⁇ standard error of the mean (s.e.m.).
  • CRI MaestroTM non-invasive fluorescence imaging system was used to follow tumor progression of mice bearing mCherry-labeled MG-63-Ras human osteosarcoma tumors and for biodistribution studies of FITC-labeled HPMA copolymer-ALN-TNP- 470 conjugate.
  • Mice were maintained on a non-fluorescent diet (Harlan) for the whole period of the experiment.
  • Mice were anesthetized using ketamine (100 mg/kg) and xylazine (12 mg/kg), treated with a depilatory cream (Veet®) and placed inside the imaging system.
  • Veet® depilatory cream
  • selected organs from mice were dissected and placed inside the imaging system.
  • Multispectral image-cubes were acquired through 550-800 nm spectral range in 10 nm steps using excitation (575-605 nni) and emission (645 ran longpass) filter set. Mice autofluorescence and undesired background signals were eliminated by spectral analysis and linear unmixing algorithm. Additionally, dissected tumors were fixed in 4 % PFA and imaged as whole-mount by confocal microscopy as described earlier in order to assess the conjugate accumulation in the tumor site.
  • Immunohistochemistry Immunohistochemistry of tumor nodules was performed using 5 ⁇ m thick formalin-fixed, paraffin-embedded tissue sections.
  • Paraffin sections were de- paraffinized, rehydrated, and stained by hematoxylin and eosin (H & E).
  • H & E hematoxylin and eosin
  • slides were deparaffinized and pre-treated with 10 niM citrate, pH 6.0 for 50 min in a steam pressure cooker (Decloaking Chamber, BioCare Medical, Walnut Creek, CA). All further steps were performed at room temperature in a hydrated chamber. Slides were covered with Peroxidase Block (Merck, Germany) for 5 minutes to quench endogenous peroxidase activity, followed by incubation with 10 % of rabbit serum in 50 mM Tris-HCl, pH 7.4, for 30 min to block non-specific binding sites.
  • Peroxidase Block Merck, Germany
  • HUVECs migration and capillary-like tube formation expressed as mean ⁇ standard deviation (s.d.).
  • In vivo data of Miles assay and evaluation of antitumor activity of HPMA copolymer-ALN-TNP-470 conjugate was expressed as mean ⁇ standard error of the mean (s.e.m.).
  • Statistical significance was determined using an unpaired Mest. P ⁇ 0.05 was considered statistically significant. All statistical tests were two-sided.
  • HPMA-Gly-Gly-Pro-Nle-ALN-TNP-470 conjugate was prepared in a two-step synthesis.
  • an intermediate was synthesized by copolymerization of HPMA, ALN- methacrylamide monomer (MA-Gly-Gly-Pro-Nle-ALN), and amino group-containing methacrylamide monomer (MA-Gly-Gly-Pro-Nle-ethylenediamine).
  • FITC fluorescein isothiocyanate
  • MA-FITC N-methacryloylaminopropyl fluorescein thiourea
  • TNP-470 was linked to free amino groups by nucleophilic substitution of the terminal chlorine of TNP-470.
  • Gly-Gly-Pro-Nle was synthesized by solid phase peptide synthesis (SPPS) and manual Fmoc/tBu strategy using 2 grams of 2-chlorotrityl chloride beads with 80 % loading, yielding the desired monomer (0.88 grams, 95 % purity).
  • SPPS solid phase peptide synthesis
  • MA-Gly-Gly-Pro-Nle-OH 100 mg, 0.24 mmol
  • 2-mercaptothiazoline TT, 33 mg, 0.28 mmol
  • MA-GIy -GIy-P ro-Nle- NH(CH 2 ) 2NH 2 MA-Gly-Gly-Pro-Nle-ethylenediamine was synthesized by SPPS using 1.5 gram of 2-chlorotrityl chloride beads. Six-time excess of ethyl enediamine in anhydrous tetrahydrofuran (THF) was applied, followed by Fmoc-amino acids, and capping with MA-GIy-GIy-OH.
  • THF anhydrous tetrahydrofuran
  • the final peptide was cleaved by 5 % trifluoroacetic acid (TFA) in dichloromethane (DCM), yielding 0.85 gram of the desired compound, in 89 % purity, as determined by HPLC (buffer A: H 2 O, 0.1 % TFA; buffer B: acetonitrile, 0.1 % TFA; gradient method: buffer B 2-60 %/30 minutes; 1 ml/minute; single peak, elution time 8.27 minutes).
  • TFA trifluoroacetic acid
  • DCM dichloromethane
  • DIPEA diisopropylethylamine
  • Nle-ethylene-NH 2 for conjugating TNP-470
  • HPMA and MA-FITC are dissolved in water in the presence of 4,4'-azobis(4-cyanovaleric acid) (VA-501) as a co- polymerization initiator.
  • VA-501 4,4'-azobis(4-cyanovaleric acid)
  • the solution is bubbled with nitrogen for 10 minutes, the ampoule is sealed, and copolymerization is performed at 60 0 C.
  • the relative amounts of the various monomeric units can be varied as desired, so as to determine the load of the alendronate, the TNP-470 and the PITC (if present) in the formed polymer.
  • the reaction conditions can further be manipulated, so as to determine the degree of polymerization or to incorporate other moieties such as tyrosine groups in order to radiolabel the conjugate
  • MA-Gly-Gly-Pro-Nle-ALN 73 mg
  • MA-Gly-Gly-Pro-Nle-ethylene-NH 2 55 mg
  • HPMA 200 mg
  • MA-FITC 4 mg, if present
  • 4,4'- azobis(4-cyanovaleric acid) VA-501, 3 mg
  • Nle-ethylene-NH 2 55 mg
  • HPMA 200 mg
  • VA-501 4,4'-azobis(4-cyanovaleric acid)
  • VA-501 3 mg
  • the solution was bubbled with nitrogen for 10 minutes, the ampoule sealed, and the mixture polymerized at 60 0 C for 24 hours.
  • RAFT reversible addition-fragmentation chain transfer
  • MA-Gly-Gly-Pro-Nle-ALN, MA-Gly-Gly-Pro-Nle-NH(CH 2 ) 2 NH 2 , HPMA, and MA-FITC are dissolved in water in the presence of 2,2'-Azobis[2-(2- imidazolin-2-yl)propane]dihydrochloride (VA-044) as an initiator and SV_»"-bis( ⁇ , ⁇ ' ⁇ dimethyl- ⁇ "-acetic acid) trithiocarbonate as a chain transfer agent (TTC).
  • VA-044 2,2'-Azobis[2-(2- imidazolin-2-yl)propane]dihydrochloride
  • SV_ "-bis( ⁇ , ⁇ ' ⁇ dimethyl- ⁇ "-acetic acid) trithiocarbonate
  • TTC chain transfer agent
  • MA-Gly-Gly-Pro-Nle-ALN (293 rag), MA-Gly-Gly-Pro-Nle- NH(CHo) 2 NH 2 (215 mg), HPMA (948 mg), MA-FITC (4 mg if present), 2,2'-Azobis[2- (2-imidazolin-2-yl)propane]dihydrochloride (VA-044 1.6 mg) as initiator and 5,5"- bis( ⁇ , ⁇ '-dimethyl- ⁇ "-acetic acid) trithiocarbonate as chain transfer agent (TTC 4.2 mg) were dissolved in 7.5 ml of water. The solution was bubbled with nitrogen for 30 minutes, sealed in ampoule, and the mixture polymerized at 30 0 C for 48 hours.
  • Both polymers were purified by dissolving in water and precipitating into an excess of acetone (3 times); following each precipitation, the precipitate was washed with acetone. Finally, the polymers were dissolved in 15 ml of water; pH adjusted to 12 with 1 N NaOH, and dialyzed against DI water for 24 hours at 4 0 C (MWCO 12-14 kDa) to remove excess ALN monomer. The sample was freeze-dried after dialysis.
  • the content of ALN, FITC and amine in the HPMA conjugate was estimated as described in the methods section hereinabove.
  • the content of ALN was determined to be 0.42 mmol/gram (7.0 mol %)
  • the content of FITC was determined to be 0.04 mmol/gram (0.6 mol %)
  • the content of amine was determined to be 0.24 mmol/gram (4.3 mol %).
  • the content of ALN was determined to be 0.18 mmol/gram (3.2 mol %), and the content of amine was determined to be 0.36 mmol/gram (6.4 mol %).
  • the content of ALN was determined to be 7.7 mol %
  • the content of FITC was determined to be 0.1 mol %
  • the content of amine was determined to be 6.3 mol %. It is therefore shown that in any of synthetic approaches described herein, the load of alendronate can be controlled as desired and further, a relatively high load can be obtained.
  • HPMA-ALN-TNP-470 conjugate HPMA copolymer-ALN-NH 2 (150 mg) was dissolved in 6 ml of dimethylformamide (DMF; if necessary, a small amount of water was added to dissolve the polymer) and the solution was cooled to 4 0 C. Then, TNP-470 (150 mg) in 1 ml DMF was added. The reaction mixture was stirred at 4 0 C in the dark for 12 hours. The conjugate was thereafter precipitated into acetone and purified by reprecipitation (3 times) from an aqueous solution into an excess of acetone. The precipitate was washed with acetone and the residue was dissolved in water and dialyzed for 1 day at 4 0 C (MWCO 12-14 kDa) against DI water. The product was isolated by free-drying.
  • DMF dimethylformamide
  • HPMA -ALN-TNP-470 conjugate eluted as a single peak with a retention time of 20 minutes as evaluated by size exclusion chromatography (SEC) (see, Figure 3B).
  • HPMA-ALN-TNP-470 conjugate size distribution The molecular weight and polydispersity of the two conjugates synthesized, namely, the conjugate polymerized by the classical polymerization method (polymerization I conjugate) and the conjugate polymerized by "living polymerization” RAFT (polymerization II conjugate), were estimated by SEC exhibiting an apparent M w of 80 kDa ( Figure 3D and 3E respectively). Additionally, the hydrodynamic diameter size distribution of the HPMA copolymer-ALN-TNP-470 conjugates was determined using an optical analyzer.
  • the conjugate polymerized by the classical polymerization method had a PDI of ⁇ 1.62 with a mean size distribution of 241 nm whereas the conjugate polymerized by "living polymerization" RAFT was well-dispersed exhibiting a considerably lower and narrower PDI of ⁇ 1.2 with a mean size distribution of 100 nm ( Figure 3F).
  • Combination treatments I cardiac concentrations of ALN and TNP-470 0.01 pM
  • II cardiac concentrations of TNP-470 and ALN 10 ⁇ M
  • HUVEC proliferation at IC 30 so, 70 of 0.2, 10, 30 ⁇ M and 0.00001, 0.004, 40 nM, respectively.
  • data from combination treatments were calculated according to CI equation and were used to generate isobolograms at IC 3 O, 50 , 70 of HUVEC proliferation by ALN-TNP-470 combinations.
  • combination treatment I had synergistic inhibitory effect on HUVEC at IC 30 , so, 70 with CI of 0.055, 0.3, and 0.S9.
  • Combination treatment II had synergistic effect at IC so, 70 with CI of 0.23, 0.121 and additive effect at IC 30 with CI of 1.025.
  • Intracellular trafficking of FITC-labeled HPMA -ALN-TNP-470 conjugate in endothelial and Saos-2 human osteosarcoma cells Following chemical characterization, the ability of FITC-labeled HPMA copolymer ALN-TNP-470 conjugate to internalize into endothelial and human osteosarcoma cells and the mechanism by which it internalizes was studied.
  • HUVEC and Saos-2 osteosarcoma cells were incubated with the conjugate, fixed, permeabilzed and stained with the nuclei marker propidium iodide (PI). Confocal microscopy was performed by separately multi-channel tracking for PI (red) and FITC-labeled conjugate (green).
  • the conjugate was capable of internalizing into HUVAC and Saos-2 cells as demonstrated by colocalization of 82 % of the conjugate with transferrin in HUVEC cells ( Figures 4L-O) and of 71 % in Saos-2 cells ( Figures 4P-S). These high percentages of colocalization suggest a lysosomotropic pathway of cellular uptake via clathrin-coated vesicles.
  • HPMA -ALN-TNP-470 conjugate Effect of HPMA -ALN-TNP-470 conjugate on proliferation of endothelial, Saos-2 and MG-63-Ras human osteosarcoma cells
  • HPMA copolymer ALN-TNP-470 conjugate is active in vitro and that the bound drugs retained their antitumor and anti-angiogenic activity following polymer-conjugation
  • the inhibitory effect of the conjugate on HUVEC, Saos- 2 and MG-63-Ras human osteosarcoma cell proliferation was examined.
  • Saos-2 human osteosarcoma cell proliferation was inhibited similarly by free and conjugated ALN and TNP-470 combinations at IC 50 of 30 ⁇ M.
  • MG-63-Ras human osteosarcoma cell proliferation was inhibited similarly by free and conjugated ALN-TNP-470 at IC 50 of 10 ⁇ M.
  • HPMA alone was inert in vitro and in vivo (data not shown), in agreement with previously published data [Duncan et al. J Control Release 2001; 74: 135-146].
  • HPMA copolymer-ALN-TNP-470 conjugate is active mainly upon the release of ALN and TNP-470 by cathepsin K cleavage mechanism
  • the inhibition of HUVEC proliferation by HPMA copolymer-ALN-TNP-470 conjugate in the presence of cathepsin K inhibitor III was evaluated.
  • HPMA copolymer-ALN-TNP- 470 conjugate inhibited the proliferation of HUVEC at a 4-logs higher concentration in the presence of cathepsin K inhibitor III than in its absence.
  • HPMA copolymer ALN-TNP-470 conjugate on vascular endothelial growth factor (VEGF)-induced HUVEC migration was examined. Migration was assessed by counting the number of cells that migrated through the membranes towards the chemoattractant VEGF during a 4 hour period following 4 hours of treatment with a combination of free ALN + free TNP-470 or conjugated HPMA-ALN-TNP-470.
  • VEGF vascular endothelial growth factor
  • HPMA copolymer-ALN-TNP-470 conjugate to inhibit capillary-like tube formation of HUVEC was examined.
  • HPMA copolymer-ALN-TNP-470 conjugate is able to reduce microvessel hyperpermeability a modified Miles assay was used. Evans blue dye was injected to the retro-orbital plexus and immediately thereafter the vascular permeability-induced factor VEGF was injected into the shaved flank of Balb/c mice.
  • Evans blue binds to plasma proteins and therefore extravasates along with them at sites of increased permeability.
  • VEGF-induced extravasation of Evans blue dye was remarkably inhibited in mice treated with free combination of ALN and TNP-470 and with HPMA copolymer-ALN-TNP-470 conjugate compared to vehicle treated mice by
  • HPMA copolymer-ALN-TNP-470 conjugate inhibits MG-63-Ras human osteosarcoma in vivo
  • HPMA copolymer- ALN-TNP-470 conjugate showed greater intensity of FITC- fluorescence spectrum (composed images of unmixed multispectral cubes) in bone tissues then in the spleen, heart, lungs, kidneys and liver ( Figure 6G). Some fluorescence is shown in the kidneys due to renal excretion of the conjugate.
  • HPMA copolymer-TNP-470 conjugates with octa-D-aspartate as a bone targeting moiety are described herein (polymers denoted as HPl and HP2).
  • the samples containing aspartate were prepared using different polymer precursors, obtained either by "thermo" polymerization (AIBN as the initiator; HPl) or "photo” polymerization (HP2).
  • the molecular weight profiles of these two polymers are different.
  • FD8 was synthesized by solid phase peptide synthesis (SPPS) using 1 gram of 2- chlorotrityl chloride beads with 50 % loading.
  • the C6 spacer (1 amino hexanoic acid) was introduced to reduce the steric hindrance during the synthesis and the following binding reaction.
  • Synthesis of polymer precursor HPMA containing Tcp and Fmoc protected amino groups [HPMA-OTcp-NH-Fmoc (Compound 4)J:
  • HPMA-OTcp-NH-Fmoc was also synthesized by photo-polymerization, as follows: 2,2-Dimethoxy-2-phenylacetophenone (DMPAP; 40 mM), instead of AIBN, was used as the initiator.
  • DMPAP 2,2-Dimethoxy-2-phenylacetophenone
  • the polymerization was conducted under light (about 1000 lm/m 2 ) for 24 hours at room temperature. Purification was performed as described hereinabove for thermo-polymerization.
  • HPMA-FDS-NH-Fmoc (Compound S)J HPMA-OTcp-NH-Fmoc (compound 4; 200 mg) was dissolved in 2.5 ml DMSO.
  • HPMA-FD8-NH-Fmoc (Compound 5; 150 mg) was dissolved in DMF (5 ml) and piperidine (1.2 ml) was added. The reaction mixture was stirred for 1 hour at room temperature. Then, the reaction mixture was acidified with acetic acid and precipitated into acetone (the polymer did not precipitate into acetone without acidifying). The copolymer was purified by washing 3 times with CHCl 3 to remove the piperidine-acetic acid salt, re-precipitated 3 times from methanol into acetone, and dried under vacuum. The polymer was further purified by FPLC (acetate buffer, pH 5.5).
  • the polymer fraction was concentrated and pH adjusted to about 8 by NaHCO 3 , followed by dialysis against DI water for 24 hours at 4 0 C (MWCO 6-8 kDa) to remove the salt.
  • the sample was freeze-dried after dialysis.
  • HPMA-FDS-NH 2 100 mg was dissolved in DMF (3 ml) and cooled to 4 0 C.
  • TNP-470 100 mg in 1 ml of DMF was added.
  • the reaction mixture was stirred at 4 0 C in dark for 12 hours.
  • the conjugate was thereafter precipitated into acetone, and purified by dissolving in water and precipitating in acetone for 3 times; after each precipitation the precipitate was washed with acetone.
  • the conjugate was further purified by FPLC (acetate buffer, pH 5.5), dialyzed for 1 day at 4 0 C (MWCO 6-8 kDa) and freeze-dried yielding the desired HPMA-D-Asp 8 -TNP- 470 conjugate (Compound 6).
  • Table 1 presents the chemical characteristics of the two HPMA-D-ASPs-TNP- 470 conjugates (HPl and HP2), prepared as described in Example 10 hereinabove, with respect to the Asp 8 content, Fmoc (implying TNP470 content) and MW.
  • the molecular weight of the conjugate was determined by size exclusion chromatography (SEC) (see, Figure 9) on AKTA/FPLC system (Pharmacia), using Superose 6 HR 10/30 column, buffer 0.1 M acetate/30 % acetonitrile, pH 5.5.
  • TNP-470 when bound to HPMA copolymer, retained its antiangiogenic effect, proliferation and migration assays were performed.
  • the proliferation of HUVEC was inhibited similarly by free TNP-470 and conjugated TNP-470 (see, Figure 10).
  • the effect of HPl and HP2 conjugates on vascular endothelial growth factor (VEGF)-induced HUVEC migration was examined. Migration was assessed by counting the number of cells that migrated through the membranes towards the chemoattractant VEGF during a 4 hour period following 4 hours of treatment with free TNP-470 or conjugated TNP-470.
  • VEGF vascular endothelial growth factor
  • Alendronate inhibits HUVEC andSaos-2 human sarcoma cells proliferation in a dose dependent manner To determine whether the anti-angiogenic activity of alendronate is dose dependent, the inhibitory effect of the drug on HUVEC and Saos-2 human osteosarcoma cell proliferation was examined.

Landscapes

  • Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biomedical Technology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Oncology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

L'invention concerne des conjugués de copolymères dérivés de l'hydroxypropylméthacrylamide (HPMA) sur lesquels sont fixés un agent TNP-470 et une charge élevée (p. ex. plus de 3 % en moles) d'alendronate (ALN), ainsi que des procédés de préparation de ces conjugués. L'invention concerne en outre des conjugués de polymères ou de copolymères sur lesquels sont fixés un agent anti-angiogenèse et un agent ciblant les os, l'oligoaspartate, ainsi que des procédés de préparation de ces conjugués. L'invention concerne également des compositions pharmaceutiques contenant ces conjugués et des utilisations de ceux-ci pour traiter et contrôler des troubles osseux.
PCT/IL2009/000511 2008-05-22 2009-05-21 Conjugué d'un polymère, d'un agent anti-angiogenèse et d'une fraction de ciblage et utilisations de ce conjugué pour traiter des maladies angiogéniques osseuses WO2009141827A2 (fr)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US12/993,855 US8703114B2 (en) 2008-05-22 2009-05-21 Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions
CN200980128588.6A CN102105157B (zh) 2008-05-22 2009-05-21 聚合物、抗血管生成剂和靶向部分的缀合物及其在制备用于治疗骨相关血管生成状况的药物中的用途
EP09750279.3A EP2300021A4 (fr) 2008-05-22 2009-05-21 Conjugué d'un polymère, d'un agent anti-angiogenèse et d'une fraction de ciblage et utilisations de ce conjugué pour traiter des maladies angiogéniques osseuses
US14/242,901 US20140212357A1 (en) 2008-05-22 2014-04-02 Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US7188808P 2008-05-22 2008-05-22
US61/071,888 2008-05-22
US19313608P 2008-10-30 2008-10-30
US61/193,136 2008-10-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US12/993,855 A-371-Of-International US8703114B2 (en) 2008-05-22 2009-05-21 Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions
US14/242,901 Continuation US20140212357A1 (en) 2008-05-22 2014-04-02 Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions

Publications (2)

Publication Number Publication Date
WO2009141827A2 true WO2009141827A2 (fr) 2009-11-26
WO2009141827A3 WO2009141827A3 (fr) 2010-01-14

Family

ID=41340640

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2009/000511 WO2009141827A2 (fr) 2008-05-22 2009-05-21 Conjugué d'un polymère, d'un agent anti-angiogenèse et d'une fraction de ciblage et utilisations de ce conjugué pour traiter des maladies angiogéniques osseuses

Country Status (4)

Country Link
US (2) US8703114B2 (fr)
EP (1) EP2300021A4 (fr)
CN (1) CN102105157B (fr)
WO (1) WO2009141827A2 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012153297A1 (fr) 2011-05-11 2012-11-15 Ramot At Tel-Aviv University Ltd. Conjugués polymères ciblés et leurs utilisations
WO2013132485A1 (fr) 2012-03-05 2013-09-12 Ramot At Tel-Aviv University Ltd. Polymères ayant des agents actifs au niveau thérapeutique conjugués à ceux-ci, procédés de préparation desdits polymères et leurs utilisations
US8586019B2 (en) 2008-05-22 2013-11-19 Ramot At Tel-Aviv University Ltd. Conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases
US8658149B2 (en) 2008-05-22 2014-02-25 Ramot At Tel-Aviv University Ltd. Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
US8703114B2 (en) 2008-05-22 2014-04-22 Ramot At Tel-Aviv University Ltd. Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions
CN103923256A (zh) * 2014-04-11 2014-07-16 西北师范大学 叶酸-苯甲醛氮芥-hpma高分子共聚物及其制备和应用
WO2017145164A1 (fr) 2016-02-24 2017-08-31 Ramot At Tel-Aviv University Ltd. Conjugués polymères et leurs utilisations
JP2019202999A (ja) * 2013-10-03 2019-11-28 スリーエム イノベイティブ プロパティズ カンパニー 結合能力が向上したリガンド官能化基材

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2544701B1 (fr) 2010-03-08 2019-08-14 University of Utah Research Foundation Conjugués d'administration de médicaments polymères, leurs procédés de fabrication et leurs méthodes d'utilisation
US9889225B2 (en) * 2010-08-20 2018-02-13 Kaohsiung Medical University Method for bone formation by administering poly(lactic-co-glycolic acid) cross-linked alendronate
RU2631486C1 (ru) * 2016-06-17 2017-09-22 Федеральное бюджетное учреждение науки "Государственный научный центр вирусологии и биотехнологии "Вектор" (ФБУН ГНЦ ВБ "Вектор") Способ получения противоопухолевого средства против метастазов костей в виде конъюгата белка-цитокина и аминобисфосфоната
US20190358341A1 (en) * 2016-12-23 2019-11-28 Commonwealth Scientific And Industrial Research Organisation Biocompatible and hydrophilic polymer conjugate for targeted delivery of an agent
WO2019060298A1 (fr) 2017-09-19 2019-03-28 Neuroenhancement Lab, LLC Procédé et appareil de neuro-activation
US11717686B2 (en) 2017-12-04 2023-08-08 Neuroenhancement Lab, LLC Method and apparatus for neuroenhancement to facilitate learning and performance
US11478603B2 (en) 2017-12-31 2022-10-25 Neuroenhancement Lab, LLC Method and apparatus for neuroenhancement to enhance emotional response
US11364361B2 (en) 2018-04-20 2022-06-21 Neuroenhancement Lab, LLC System and method for inducing sleep by transplanting mental states
WO2020056418A1 (fr) 2018-09-14 2020-03-19 Neuroenhancement Lab, LLC Système et procédé d'amélioration du sommeil
CN109289054B (zh) * 2018-09-30 2021-12-28 中南大学湘雅医院 一种特异靶向骨组织的plga-peg-zol载药纳米材料及其制备方法
US11786694B2 (en) 2019-05-24 2023-10-17 NeuroLight, Inc. Device, method, and app for facilitating sleep

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003086382A1 (fr) 2002-04-11 2003-10-23 Children's Medical Center Corporation Conjugues de polymeres tnp-470 et utilisation de ceux-ci
WO2003086178A2 (fr) 2002-04-11 2003-10-23 Children's Medical Center Corporation Methodes permettant d'inhiber l'hyperpermeabilite vasculaire
WO2004062588A2 (fr) 2003-01-06 2004-07-29 University Of Utah Systeme de liberation de medicament polymerique soluble dans l'eau ciblant les os
US6884817B2 (en) 1996-03-12 2005-04-26 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MXPA02007939A (es) * 2000-02-24 2003-02-10 Genentech Inc Terapia con profarmacos activados por la caspasa.
EP1610751A4 (fr) * 2001-04-26 2006-05-24 Univ Texas Compositions therapeutiques conjuguees agent/ligand, leur methodes de synthese et leur utilisation
US20030217320A1 (en) * 2002-05-20 2003-11-20 Gorshe Steven Scott Cyclic redundancy check circuit for use with self-synchronous scramblers
DE10310082A1 (de) * 2003-03-07 2004-09-16 Ktb Tumorforschungsgesellschaft Mbh Protein-bindende Doxorubicin-Peptid-Derivate
CA2572226A1 (fr) 2004-06-28 2006-02-02 Bruce R. Line Nanohybrides radiomarques ciblant un systeme neovasculaire de la tumeur solide et procede d'utilisation associe
WO2006084054A2 (fr) 2005-02-02 2006-08-10 Children's Medical Center Corporation Procede de traitement de maladies angiogeniques
US8404650B2 (en) * 2005-10-28 2013-03-26 The Regents Of The University Of Colorado, A Body Corporate Methods of treating cancer with doxazolidine and prodrugs thereof
US8252740B2 (en) 2006-01-27 2012-08-28 The University Of Mississippi Medical Center Thermally-targeted delivery of medicaments including doxorubicin
CA2663375A1 (fr) * 2006-09-15 2008-03-20 Onconova Therapeutics, Inc. Composes cytotoxiques actives destines a se fixer a des molecules de ciblage dans le traitement de maladies touchant des mammiferes
WO2008141110A2 (fr) * 2007-05-09 2008-11-20 Nitto Denko Corporation Conjugués de polyglutamate et conjugués de polyglutamate-amino acide contenant une pluralité de médicaments
WO2009141826A2 (fr) 2008-05-22 2009-11-26 Ramot At Tel Aviv University Ltd. Nouveaux conjugués de polymères sur lesquels sont fixés un agent thérapeutiquement actif et une fraction ciblant l'angiogenèse et utilisations de ces conjugués pour traiter des maladies liées à l'angiogenèse
WO2009141827A2 (fr) 2008-05-22 2009-11-26 Ramot At Tel Aviv University Ltd. Conjugué d'un polymère, d'un agent anti-angiogenèse et d'une fraction de ciblage et utilisations de ce conjugué pour traiter des maladies angiogéniques osseuses
CN102131509B (zh) 2008-05-22 2015-01-07 特拉维夫大学拉莫特有限公司 聚合物、二膦酸盐和抗血管生成剂的缀合物及其在治疗和监测骨相关疾病中的用途
WO2013132485A1 (fr) 2012-03-05 2013-09-12 Ramot At Tel-Aviv University Ltd. Polymères ayant des agents actifs au niveau thérapeutique conjugués à ceux-ci, procédés de préparation desdits polymères et leurs utilisations

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6884817B2 (en) 1996-03-12 2005-04-26 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
WO2003086382A1 (fr) 2002-04-11 2003-10-23 Children's Medical Center Corporation Conjugues de polymeres tnp-470 et utilisation de ceux-ci
WO2003086178A2 (fr) 2002-04-11 2003-10-23 Children's Medical Center Corporation Methodes permettant d'inhiber l'hyperpermeabilite vasculaire
WO2004062588A2 (fr) 2003-01-06 2004-07-29 University Of Utah Systeme de liberation de medicament polymerique soluble dans l'eau ciblant les os

Non-Patent Citations (31)

* Cited by examiner, † Cited by third party
Title
BERGE ET AL.: "Pharmaceutical Salts", JOURNAL OF PHARMACEUTICAL SCIENCE, vol. 66, 1977, pages 1 - 19, XP002675560, DOI: doi:10.1002/jps.2600660104
CABEBE ET AL., CURR TREAT OPTIONS ONCOL, vol. 8, 2007, pages 15 - 27
CHENG ET AL., PEDIATR BLOOD CANCER, vol. 42, 2004, pages 410 - 415
CHOU T.C., PHARMOCOL REV, vol. 58, 2006, pages 621 - 681
CLAFFEY ET AL., CANCER RES, vol. 56, 1996, pages 172 - 181
DUNCAN ET AL., J CONTROL RELEASE, vol. 74, 2001, pages 135 - 146
DUNCAN, J CONTROL RELEASE, vol. 74, 2001, pages 135 - 146
FINGL ET AL.: "The Pharmacological Basis of Therapeutics", 1975, pages: L
FOLKMAN, J. APMIS, vol. 112, 2004, pages 496 - 507
HASHIMOTO ET AL., BIOCHEM BIPHYS RES COMMUN, vol. 354, 2007, pages 478 - 484
HASHIMOTO ET AL., CANCER RES, vol. 65, 2005, pages 540 - 545
HRUBY ET AL., JOURNAL OF APPLIED POLYMER SCIENCE, vol. 101, 2006, pages 3192 - 3201
HURWITZ ET AL., N ENGL J MED, vol. 350, 2004, pages 2335 - 2342
HUSMANN ET AL., MOL CARCINOG, vol. 47, 2008, pages 66 - 73
LARRIVEE ET AL., METHODS MOL BIOL, vol. 290, 2005, pages 315 - 329
MILES; MILES, J PHYSIOL, vol. 118, 1952, pages 228 - 257
MOLINUEVO ET AL., EUR J PHARMACOL, vol. 562, 2007, pages 28 - 33
O'HARE ET AL., JOURNAL OF DRUG TARGETING, vol. 1, 1993, pages 217 - 229
PAN ET AL., J DRUG TARGET, vol. 14, 2006, pages 425 - 435
QUAN ET AL., CANCER METASTASIS REV, vol. 10, 2006, pages 707 - 713
RUSSELL RG, PEDIATRICS, vol. 119, no. 2, 2007, pages 150 - 162
SATCHI-FAINARO ET AL., CANCER CELL, vol. 7, 2005, pages 251 - 261
SATCHI-FAINARO ET AL., NAT MED, vol. 10, 2004, pages 255 - 261
See also references of EP2300021A4
SEYMOUR ET AL., JOURNAL OF CLINICAL ONCOLOGY, vol. 20, 2002, pages 1668 - 1676
TERWOGT ET AL., ANTICANCER DRUGS, vol. 12, 2001, pages 315 - 323
TUOMELA ET AL., BMC CANCER, vol. 8, 2008, pages 81
WANG ET AL., BIOCONJUG CHEM, vol. 14, 2003, pages 853 - 859
WANG ET AL., MOL PHARM, vol. 3, 2006, pages 717 - 725
WEIDNER ET AL., NENGL JMED, vol. 324, 1991, pages 1 - 8
WOOD ET AL., J PHARMACOL EXP THER, vol. 302, 2002, pages 1055 - 1061

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9259482B2 (en) 2008-05-22 2016-02-16 Ramot At Tel-Aviv University Ltd. Conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases
US9095618B2 (en) 2008-05-22 2015-08-04 Ramot At Tel-Aviv University Ltd. Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
US8586019B2 (en) 2008-05-22 2013-11-19 Ramot At Tel-Aviv University Ltd. Conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases
US8658149B2 (en) 2008-05-22 2014-02-25 Ramot At Tel-Aviv University Ltd. Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
US9427474B2 (en) 2008-05-22 2016-08-30 Ramot At Tel-Aviv University Ltd. Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
US8703114B2 (en) 2008-05-22 2014-04-22 Ramot At Tel-Aviv University Ltd. Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions
WO2012153297A1 (fr) 2011-05-11 2012-11-15 Ramot At Tel-Aviv University Ltd. Conjugués polymères ciblés et leurs utilisations
WO2013132485A1 (fr) 2012-03-05 2013-09-12 Ramot At Tel-Aviv University Ltd. Polymères ayant des agents actifs au niveau thérapeutique conjugués à ceux-ci, procédés de préparation desdits polymères et leurs utilisations
US9687562B2 (en) 2012-03-05 2017-06-27 Ramot At Tel-Aviv University Ltd. Polymers having therapeutically active agents conjugated thereto, processes of preparing same and uses thereof
US10155047B2 (en) 2012-03-05 2018-12-18 Ramot At Tel-Aviv University Ltd. Polymers having therapeutically active agents conjugated thereto, processes of preparing same and uses thereof
JP2019202999A (ja) * 2013-10-03 2019-11-28 スリーエム イノベイティブ プロパティズ カンパニー 結合能力が向上したリガンド官能化基材
US11237085B2 (en) 2013-10-03 2022-02-01 3M Innovative Properties Company Ligand-functionalized substrates with enhanced binding capacity
CN103923256A (zh) * 2014-04-11 2014-07-16 西北师范大学 叶酸-苯甲醛氮芥-hpma高分子共聚物及其制备和应用
CN103923256B (zh) * 2014-04-11 2015-11-04 西北师范大学 叶酸-苯甲醛氮芥-hpma高分子共聚物及其制备和应用
WO2017145164A1 (fr) 2016-02-24 2017-08-31 Ramot At Tel-Aviv University Ltd. Conjugués polymères et leurs utilisations
US11666656B2 (en) 2016-02-24 2023-06-06 Ramot At Tel-Aviv University Ltd. Polymeric conjugates and uses thereof

Also Published As

Publication number Publication date
EP2300021A4 (fr) 2014-10-08
US20140212357A1 (en) 2014-07-31
US8703114B2 (en) 2014-04-22
WO2009141827A3 (fr) 2010-01-14
US20110085979A1 (en) 2011-04-14
EP2300021A2 (fr) 2011-03-30
CN102105157A (zh) 2011-06-22
CN102105157B (zh) 2015-09-30

Similar Documents

Publication Publication Date Title
US8703114B2 (en) Conjugate of a polymer, an anti-angiogenesis agent and a targeting moiety, and uses thereof in the treatment of bone related angiogenesis conditions
US9427474B2 (en) Conjugates of a polymer, a bisphosphonate and an anti-angiogenesis agent and uses thereof in the treatment and monitoring of bone related diseases
US10155047B2 (en) Polymers having therapeutically active agents conjugated thereto, processes of preparing same and uses thereof
US9283279B2 (en) Targeted polymeric conjugates and uses thereof
US9259482B2 (en) Conjugates of polymers having a therapeutically active agent and an angiogenesis targeting moiety attached thereto and uses thereof in the treatment of angiogenesis related diseases
Eldar-Boock et al. Integrin-assisted drug delivery of nano-scaled polymer therapeutics bearing paclitaxel
US20090311182A1 (en) Macromolecular Delivery Systems for Non-Invasive Imaging, Evaluation and Treatment of Arthritis and Other Inflammatory Diseases
CN102264396A (zh) Hpma-多西他赛或吉西他滨缀合物及其用途
US20230277677A1 (en) Polymeric conjugates and uses thereof
CN113710323A (zh) 偶联物及癌治疗剂
Kern Polymerizable Peptide Monomers for the Targeted and Intracellular Delivery of Cancer Therapeutics
Zhang et al. Poly (L-glutamic acid): efficient carrier of cancer therapeutics and diagnostics

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200980128588.6

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09750279

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 12993855

Country of ref document: US

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2558/MUMNP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2009750279

Country of ref document: EP